1
|
Hill BM, Holloway RK, Forbes LH, Davies CL, Monteiro JK, Brown CM, Rose J, Fudge N, Plant PJ, Mahmood A, Brand-Arzamendi K, Kent SA, Molina-Gonzalez I, Gyoneva S, Ransohoff RM, Wipke B, Priller J, Schneider R, Moore CS, Miron VE. Monocyte-secreted Wnt reduces the efficiency of central nervous system remyelination. PLoS Biol 2025; 23:e3003073. [PMID: 40233100 PMCID: PMC12052099 DOI: 10.1371/journal.pbio.3003073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 05/05/2025] [Accepted: 02/18/2025] [Indexed: 04/17/2025] Open
Abstract
The regeneration of myelin in the central nervous system (CNS) reinstates nerve health and function, yet its decreased efficiency with aging and progression of neurodegenerative disease contributes to axonal loss and/or degeneration. Although CNS myeloid cells have been implicated in regulating the efficiency of remyelination, the distinct contribution of blood monocytes versus that of resident microglia is unclear. Here, we reveal that monocytes have non-redundant functions compared to microglia in regulating remyelination. Using a transgenic mouse in which classical monocytes have reduced egress from bone marrow (Ccr2-/-), we demonstrate that monocytes drive the timely onset of oligodendrocyte differentiation and myelin protein expression, yet impede myelin production. Ribonucleic acid sequencing revealed a Wnt signature in wild-type mouse lesion monocytes, which was confirmed in monocytes from multiple sclerosis white matter lesions and blood. Genetic or pharmacological inhibition of Wnt release by monocytes increased remyelination. Our findings reveal monocytes to be critical regulators of remyelination and identify monocytic Wnt signaling as a promising therapeutic target to inhibit for increased efficiency of CNS regeneration.
Collapse
Affiliation(s)
- Bianca M. Hill
- Keenan Research Centre for Biomedical Science of St. Michael’s Hospital, Toronto, Ontario, Canada
- BARLO Multiple Sclerosis Centre, St. Michael’s Hospital, Toronto, Ontario, Canada
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| | - Rebecca K. Holloway
- Keenan Research Centre for Biomedical Science of St. Michael’s Hospital, Toronto, Ontario, Canada
- BARLO Multiple Sclerosis Centre, St. Michael’s Hospital, Toronto, Ontario, Canada
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| | - Lindsey H. Forbes
- United Kingdom Dementia Research Institute at The University of Edinburgh, Edinburgh, Scotland, United Kingdom
- Centre for Discovery Brain Sciences, Chancellor’s Building, The University of Edinburgh, Edinburgh, Scotland, United Kingdom
| | - Claire L. Davies
- Centre for Discovery Brain Sciences, Chancellor’s Building, The University of Edinburgh, Edinburgh, Scotland, United Kingdom
| | - Jonathan K. Monteiro
- Keenan Research Centre for Biomedical Science of St. Michael’s Hospital, Toronto, Ontario, Canada
- BARLO Multiple Sclerosis Centre, St. Michael’s Hospital, Toronto, Ontario, Canada
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| | - Christina M. Brown
- United Kingdom Dementia Research Institute at The University of Edinburgh, Edinburgh, Scotland, United Kingdom
- Centre for Discovery Brain Sciences, Chancellor’s Building, The University of Edinburgh, Edinburgh, Scotland, United Kingdom
| | - Jamie Rose
- United Kingdom Dementia Research Institute at The University of Edinburgh, Edinburgh, Scotland, United Kingdom
- Centre for Discovery Brain Sciences, Chancellor’s Building, The University of Edinburgh, Edinburgh, Scotland, United Kingdom
| | - Neva Fudge
- Division of BioMedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John’s, Newfoundland, Canada
| | - Pamela J. Plant
- Keenan Research Centre for Biomedical Science of St. Michael’s Hospital, Toronto, Ontario, Canada
| | - Ayisha Mahmood
- United Kingdom Dementia Research Institute at The University of Edinburgh, Edinburgh, Scotland, United Kingdom
- Centre for Discovery Brain Sciences, Chancellor’s Building, The University of Edinburgh, Edinburgh, Scotland, United Kingdom
| | - Koroboshka Brand-Arzamendi
- Keenan Research Centre for Biomedical Science of St. Michael’s Hospital, Toronto, Ontario, Canada
- BARLO Multiple Sclerosis Centre, St. Michael’s Hospital, Toronto, Ontario, Canada
| | - Sarah A. Kent
- United Kingdom Dementia Research Institute at The University of Edinburgh, Edinburgh, Scotland, United Kingdom
- Centre for Discovery Brain Sciences, Chancellor’s Building, The University of Edinburgh, Edinburgh, Scotland, United Kingdom
| | - Irene Molina-Gonzalez
- United Kingdom Dementia Research Institute at The University of Edinburgh, Edinburgh, Scotland, United Kingdom
- Centre for Discovery Brain Sciences, Chancellor’s Building, The University of Edinburgh, Edinburgh, Scotland, United Kingdom
| | - Stefka Gyoneva
- Previously at Biogen Ltd, Cambridge, Massachusetts, United States of America
| | - Richard M. Ransohoff
- Previously at Biogen Ltd, Cambridge, Massachusetts, United States of America
- Third Rock Ventures, Boston, Massachusetts, United States of America
| | - Brian Wipke
- Previously at Biogen Ltd, Cambridge, Massachusetts, United States of America
- Manifold Bio, Boston, Massachusetts, United States of America
| | - Josef Priller
- United Kingdom Dementia Research Institute at The University of Edinburgh, Edinburgh, Scotland, United Kingdom
- Centre for Clinical Brain Sciences, Chancellor’s Building, The University of Edinburgh, Edinburgh, Scotland, United Kingdom
- Department of Psychiatry and Psychotherapy, Klinikum rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany
- Neuropsychiatry and Laboratory of Molecular Psychiatry, Charité-Universitätsmedizin Berlin and DZNE, Berlin, Germany
| | - Raphael Schneider
- Keenan Research Centre for Biomedical Science of St. Michael’s Hospital, Toronto, Ontario, Canada
- BARLO Multiple Sclerosis Centre, St. Michael’s Hospital, Toronto, Ontario, Canada
| | - Craig S. Moore
- Division of BioMedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John’s, Newfoundland, Canada
| | - Veronique E. Miron
- Keenan Research Centre for Biomedical Science of St. Michael’s Hospital, Toronto, Ontario, Canada
- BARLO Multiple Sclerosis Centre, St. Michael’s Hospital, Toronto, Ontario, Canada
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
- United Kingdom Dementia Research Institute at The University of Edinburgh, Edinburgh, Scotland, United Kingdom
- Centre for Discovery Brain Sciences, Chancellor’s Building, The University of Edinburgh, Edinburgh, Scotland, United Kingdom
| |
Collapse
|
2
|
Shang Y, Zheng L, Du Y, Shang T, Liu X, Zou W. Role of Regulatory T Cells in Intracerebral Hemorrhage. Mol Neurobiol 2025; 62:518-532. [PMID: 38877366 DOI: 10.1007/s12035-024-04281-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Accepted: 06/03/2024] [Indexed: 06/16/2024]
Abstract
Intracerebral hemorrhage (ICH) is a common cerebrovascular disease that can lead to severe neurological dysfunction in surviving patients, resulting in a heavy burden on patients and their families. When ICH occurs, the blood‒brain barrier is disrupted, thereby promoting immune cell migration into damaged brain tissue. As important immunosuppressive T cells, regulatory T (Treg) cells are involved in the maintenance of immune homeostasis and the suppression of immune responses after ICH. Treg cells mitigate brain tissue damage after ICH in a variety of ways, such as inhibiting the neuroinflammatory response, protecting against blood‒brain barrier damage, reducing oxidative stress damage and promoting nerve repair. In this review, we discuss the changes in Treg cells in ICH clinical patients and experimental animals, the mechanisms by which Treg cells regulate ICH and treatments targeting Treg cells in ICH, aiming to support new therapeutic strategies for clinical treatment.
Collapse
Affiliation(s)
- Yaxin Shang
- The Graduate School, Heilongjiang University of Chinese Medicine, Harbin, 150000, Heilongjiang, People's Republic of China
| | - Lei Zheng
- The First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, 150000, Heilongjiang, People's Republic of China
- Molecular Biology Laboratory of Clinical Integrated of Traditional Chinese and Western Medicine of Heilong Jiang Province, Heilongjiang University of Chinese Medicine, Harbin, 150000, Heilongjiang, People's Republic of China
| | - Yunpeng Du
- The Graduate School, Heilongjiang University of Chinese Medicine, Harbin, 150000, Heilongjiang, People's Republic of China
| | - Tong Shang
- The Graduate School, Heilongjiang University of Chinese Medicine, Harbin, 150000, Heilongjiang, People's Republic of China
| | - Xueting Liu
- The Graduate School, Heilongjiang University of Chinese Medicine, Harbin, 150000, Heilongjiang, People's Republic of China
| | - Wei Zou
- The First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, 150000, Heilongjiang, People's Republic of China.
- Molecular Biology Laboratory of Clinical Integrated of Traditional Chinese and Western Medicine of Heilong Jiang Province, Heilongjiang University of Chinese Medicine, Harbin, 150000, Heilongjiang, People's Republic of China.
| |
Collapse
|
3
|
Spiteri AG, Pilkington KR, Wishart CL, Macia L, King NJC. High-Dimensional Methods of Single-Cell Microglial Profiling to Enhance Understanding of Neuropathological Disease. Curr Protoc 2024; 4:e985. [PMID: 38439574 DOI: 10.1002/cpz1.985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/06/2024]
Abstract
Microglia are the innate myeloid cells of the central nervous system (CNS) parenchyma, functionally implicated in almost every defined neuroinflammatory and neurodegenerative disorder. Current understanding of disease pathogenesis for many neuropathologies is limited and/or lacks reliable diagnostic markers, vaccines, and treatments. With the increasing aging of society and rise in neurogenerative diseases, improving our understanding of their pathogenesis is essential. Analysis of microglia from murine disease models provides an investigative tool to unravel disease processes. In many neuropathologies, bone-marrow-derived monocytes are recruited to the CNS, adopting a phenotype similar to that of microglia. This significantly confounds the accurate identification of cell-type-specific functions and downstream therapeutic targeting. The increased capacity to analyze more phenotypic markers using spectral-cytometry-based technologies allows improved separation of microglia from monocyte-derived cells. Full-spectrum profiling enables enhanced marker resolution, time-efficient analysis of >40 fluorescence parameters, and extraction of cellular autofluorescence parameters. Coupling this system with additional cytometric technologies, including cell sorting and high-parameter imaging, can improve the understanding of microglial phenotypes in disease. To this end, we provide detailed, step-by-step protocols for the analysis of murine brain tissue by high-parameter ex vivo cytometric analysis using the Aurora spectral cytometer (Cytek), including best practices for unmixing and autofluorescence extraction, cell sorting for single-cell RNA analysis, and imaging mass cytometry. Together, this provides a toolkit for researchers to comprehensively investigate microglial disease processes at protein, RNA, and spatial levels for the identification of therapeutic targets in neuropathology. © 2024 The Authors. Current Protocols published by Wiley Periodicals LLC. Basic Protocol 1: Processing the mouse brain into a single-cell suspension for microglia isolation Basic Protocol 2: Staining single-cell mouse brain suspensions for microglial phenotyping by spectral cytometry Basic Protocol 3: Flow cytometric sorting of mouse microglia for ex vivo analysis Basic Protocol 4: Processing the mouse brain for imaging mass cytometry for spatial microglia analysis.
Collapse
Affiliation(s)
- Alanna G Spiteri
- Viral Immunopathology Laboratory, Infection, Immunity and Inflammation Research Theme, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
- Charles Perkins Centre, The University of Sydney, Sydney, Australia
| | | | - Claire L Wishart
- Viral Immunopathology Laboratory, Infection, Immunity and Inflammation Research Theme, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
- Charles Perkins Centre, The University of Sydney, Sydney, Australia
| | - Laurence Macia
- Charles Perkins Centre, The University of Sydney, Sydney, Australia
- Sydney Cytometry, The University of Sydney and Centenary Institute, Sydney, Australia
| | - Nicholas J C King
- Viral Immunopathology Laboratory, Infection, Immunity and Inflammation Research Theme, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
- Charles Perkins Centre, The University of Sydney, Sydney, Australia
- Sydney Cytometry, The University of Sydney and Centenary Institute, Sydney, Australia
- The University of Sydney Institute for Infectious Diseases, The University of Sydney, Sydney, Australia
- The University of Sydney Nano Institute, The University of Sydney, Sydney, Australia
| |
Collapse
|
4
|
Kent SA, Miron VE. Microglia regulation of central nervous system myelin health and regeneration. Nat Rev Immunol 2024; 24:49-63. [PMID: 37452201 DOI: 10.1038/s41577-023-00907-4] [Citation(s) in RCA: 55] [Impact Index Per Article: 55.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/13/2023] [Indexed: 07/18/2023]
Abstract
Microglia are resident macrophages of the central nervous system that have key functions in its development, homeostasis and response to damage and infection. Although microglia have been increasingly implicated in contributing to the pathology that underpins neurological dysfunction and disease, they also have crucial roles in neurological homeostasis and regeneration. This includes regulation of the maintenance and regeneration of myelin, the membrane that surrounds neuronal axons, which is required for axonal health and function. Myelin is damaged with normal ageing and in several neurodegenerative diseases, such as multiple sclerosis and Alzheimer disease. Given the lack of approved therapies targeting myelin maintenance or regeneration, it is imperative to understand the mechanisms by which microglia support and restore myelin health to identify potential therapeutic approaches. However, the mechanisms by which microglia regulate myelin loss or integrity are still being uncovered. In this Review, we discuss recent work that reveals the changes in white matter with ageing and neurodegenerative disease, how this relates to microglia dynamics during myelin damage and regeneration, and factors that influence the regenerative functions of microglia.
Collapse
Affiliation(s)
- Sarah A Kent
- UK Dementia Research Institute at The University of Edinburgh, Edinburgh, UK
- Centre for Discovery Brain Sciences, Chancellor's Building, The University of Edinburgh, Edinburgh, UK
| | - Veronique E Miron
- UK Dementia Research Institute at The University of Edinburgh, Edinburgh, UK.
- Centre for Discovery Brain Sciences, Chancellor's Building, The University of Edinburgh, Edinburgh, UK.
- Barlo Multiple Sclerosis Centre, St Michael's Hospital, Toronto, Ontario, Canada.
- Keenan Research Centre for Biomedical Science, St Michael's Hospital, Toronto, Ontario, Canada.
- Department of Immunology, The University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
5
|
Thougaard E, Carney B, Wlodarczyk A, Brambilla R, Lambertsen KL. Peripherally derived myeloid cells induce disease-dependent phenotypic changes in microglia. Front Cell Neurosci 2023; 17:1295840. [PMID: 38155863 PMCID: PMC10752942 DOI: 10.3389/fncel.2023.1295840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2023] [Accepted: 11/23/2023] [Indexed: 12/30/2023] Open
Abstract
In central nervous system (CNS) injury and disease, peripherally derived myeloid cells infiltrate the CNS parenchyma and interact with resident cells, propagating the neuroinflammatory response. Because peripheral myeloid populations differ profoundly depending on the type and phase of injury, their crosstalk with CNS resident cells, particularly microglia, will lead to different functional outcomes. Thus, understanding how peripheral myeloid cells affect the phenotype and function of microglia in different disease conditions and phases may lead to a better understanding of disease-specific targetable pathways for neuroprotection and neurorepair. To this end, we set out to develop an in vitro system to investigate the communication between peripheral myeloid cells and microglia, with the goal of uncovering potential differences due to disease type and timing. We isolated peripheral myeloid cells from mice undergoing experimental autoimmune encephalomyelitis (EAE), a model of multiple sclerosis, or acute cerebral ischemia by permanent middle cerebral artery occlusion (pMCAO) at different times after disease and probed their ability to change the phenotype of primary microglia isolated from the brain of adult mice. We identified changes not only dependent on the disease model, but also on the timepoint after disease onset from which the myeloid cells were isolated. Peripheral myeloid cells from acute EAE induced morphological changes in microglia, followed by increases in expression of genes involved in inflammatory signaling. Conversely, it was the peripheral myeloid cells from the chronic phase of pMCAO that induced gene expression changes in genes involved in inflammatory signaling and phagocytosis, which was not followed by a change in morphology. This underscores the importance of understanding the role of infiltrating myeloid cells in different disease contexts and phases. Furthermore, we showed that our assay is a valuable tool for investigating myeloid cell interactions in a range of CNS neuroinflammatory conditions.
Collapse
Affiliation(s)
- Estrid Thougaard
- Neurobiology Research, Department of Molecular Medicine, University of Southern Denmark, Odense, Denmark
- BRIDGE - Brain Research - Inter Disciplinary Guided Excellence, Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Brianna Carney
- The Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Agnieszka Wlodarczyk
- Neurobiology Research, Department of Molecular Medicine, University of Southern Denmark, Odense, Denmark
- BRIDGE - Brain Research - Inter Disciplinary Guided Excellence, Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Roberta Brambilla
- Neurobiology Research, Department of Molecular Medicine, University of Southern Denmark, Odense, Denmark
- BRIDGE - Brain Research - Inter Disciplinary Guided Excellence, Department of Clinical Research, University of Southern Denmark, Odense, Denmark
- The Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Kate Lykke Lambertsen
- Neurobiology Research, Department of Molecular Medicine, University of Southern Denmark, Odense, Denmark
- BRIDGE - Brain Research - Inter Disciplinary Guided Excellence, Department of Clinical Research, University of Southern Denmark, Odense, Denmark
- Department of Neurology, Odense University Hospital, Odense, Denmark
| |
Collapse
|
6
|
Lana D, Magni G, Landucci E, Wenk GL, Pellegrini-Giampietro DE, Giovannini MG. Phenomic Microglia Diversity as a Druggable Target in the Hippocampus in Neurodegenerative Diseases. Int J Mol Sci 2023; 24:13668. [PMID: 37761971 PMCID: PMC10531074 DOI: 10.3390/ijms241813668] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 08/31/2023] [Accepted: 09/02/2023] [Indexed: 09/29/2023] Open
Abstract
Phenomics, the complexity of microglia phenotypes and their related functions compels the continuous study of microglia in disease animal models to find druggable targets for neurodegenerative disorders. Activation of microglia was long considered detrimental for neuron survival, but more recently it has become apparent that the real scenario of microglia morphofunctional diversity is far more complex. In this review, we discuss the recent literature on the alterations in microglia phenomics in the hippocampus of animal models of normal brain aging, acute neuroinflammation, ischemia, and neurodegenerative disorders, such as AD. Microglia undergo phenomic changes consisting of transcriptional, functional, and morphological changes that transform them into cells with different properties and functions. The classical subdivision of microglia into M1 and M2, two different, all-or-nothing states is too simplistic, and does not correspond to the variety of phenotypes recently discovered in the brain. We will discuss the phenomic modifications of microglia focusing not only on the differences in microglia reactivity in the diverse models of neurodegenerative disorders, but also among different areas of the brain. For instance, in contiguous and highly interconnected regions of the rat hippocampus, microglia show a differential, finely regulated, and region-specific reactivity, demonstrating that microglia responses are not uniform, but vary significantly from area to area in response to insults. It is of great interest to verify whether the differences in microglia reactivity may explain the differential susceptibility of different brain areas to insults, and particularly the higher sensitivity of CA1 pyramidal neurons to inflammatory stimuli. Understanding the spatiotemporal heterogeneity of microglia phenomics in health and disease is of paramount importance to find new druggable targets for the development of novel microglia-targeted therapies in different CNS disorders. This will allow interventions in three different ways: (i) by suppressing the pro-inflammatory properties of microglia to limit the deleterious effect of their activation; (ii) by modulating microglia phenotypic change to favor anti-inflammatory properties; (iii) by influencing microglia priming early in the disease process.
Collapse
Affiliation(s)
- Daniele Lana
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Viale Pieraccini 6, 50139 Florence, Italy; (E.L.); (D.E.P.-G.); (M.G.G.)
| | - Giada Magni
- Institute of Applied Physics “Nello Carrara”, National Research Council (IFAC-CNR), Via Madonna del Piano 10, 50019 Florence, Italy;
| | - Elisa Landucci
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Viale Pieraccini 6, 50139 Florence, Italy; (E.L.); (D.E.P.-G.); (M.G.G.)
| | - Gary L. Wenk
- Department of Psychology, The Ohio State University, Columbus, OH 43210, USA;
| | - Domenico Edoardo Pellegrini-Giampietro
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Viale Pieraccini 6, 50139 Florence, Italy; (E.L.); (D.E.P.-G.); (M.G.G.)
| | - Maria Grazia Giovannini
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Viale Pieraccini 6, 50139 Florence, Italy; (E.L.); (D.E.P.-G.); (M.G.G.)
| |
Collapse
|
7
|
Spiteri AG, Wishart CL, Ni D, Viengkhou B, Macia L, Hofer MJ, King NJC. Temporal tracking of microglial and monocyte single-cell transcriptomics in lethal flavivirus infection. Acta Neuropathol Commun 2023; 11:60. [PMID: 37016414 PMCID: PMC10074823 DOI: 10.1186/s40478-023-01547-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Accepted: 03/08/2023] [Indexed: 04/06/2023] Open
Abstract
As the resident parenchymal myeloid population in the central nervous system (CNS), microglia are strategically positioned to respond to neurotropic virus invasion and have been implicated in promoting both disease resolution and progression in the acute and post-infectious phase of virus encephalitis. In a mouse model of West Nile virus encephalitis (WNE), infection of the CNS results in recruitment of large numbers of peripheral immune cells into the brain, the majority being nitric oxide (NO)-producing Ly6Chi inflammatory monocyte-derived cells (MCs). In this model, these cells enhance immunopathology and mortality. However, the contribution of microglia to this response is currently undefined. Here we used a combination of experimental tools, including single-cell RNA sequencing (scRNA-seq), microglia and MC depletion reagents, high-dimensional spectral cytometry and computational algorithms to dissect the differential contribution of microglia and MCs to the anti-viral immune response in severe neuroinflammation seen in WNE. Intriguingly, analysis of scRNA-seq data revealed 6 unique microglia and 3 unique MC clusters that were predominantly timepoint-specific, demonstrating substantial transcriptional adaptation with disease progression over the course of WNE. While microglia and MC adopted unique gene expression profiles, gene ontology enrichment analysis, coupled with microglia and MC depletion studies, demonstrated a role for both of these cells in the trafficking of peripheral immune cells into the CNS, T cell responses and viral clearance. Over the course of infection, microglia transitioned from a homeostatic to an anti-viral and then into an immune cell-recruiting phenotype. Conversely, MC adopted antigen-presenting, immune cell-recruiting and NO-producing phenotypes, which all had anti-viral function. Overall, this study defines for the first time the single-cell transcriptomic responses of microglia and MCs over the course of WNE, demonstrating both protective and pathological roles of these cells that could potentially be targeted for differential therapeutic intervention to dampen immune-mediated pathology, while maintaining viral clearance functions.
Collapse
Affiliation(s)
- Alanna G Spiteri
- Viral Immunopathology Laboratory, Infection, Immunity and Inflammation Research Theme, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, 2006, Australia
- Sydney Cytometry, The University of Sydney and Centenary Institute, Sydney, NSW, 2006, Australia
- Ramaciotti Facility for Human Systems Biology, The University of Sydney and Centenary Institute, Sydney, NSW, 2006, Australia
- Charles Perkins Centre, The University of Sydney, Sydney, NSW, 2006, Australia
| | - Claire L Wishart
- Viral Immunopathology Laboratory, Infection, Immunity and Inflammation Research Theme, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, 2006, Australia
- Sydney Cytometry, The University of Sydney and Centenary Institute, Sydney, NSW, 2006, Australia
- Ramaciotti Facility for Human Systems Biology, The University of Sydney and Centenary Institute, Sydney, NSW, 2006, Australia
- Charles Perkins Centre, The University of Sydney, Sydney, NSW, 2006, Australia
| | - Duan Ni
- Charles Perkins Centre, The University of Sydney, Sydney, NSW, 2006, Australia
- Chronic Diseases Research Theme, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, 2006, Australia
| | - Barney Viengkhou
- Charles Perkins Centre, The University of Sydney, Sydney, NSW, 2006, Australia
- School of Life and Environmental Sciences, The University of Sydney, Sydney, NSW, 2006, Australia
| | - Laurence Macia
- Sydney Cytometry, The University of Sydney and Centenary Institute, Sydney, NSW, 2006, Australia
- Charles Perkins Centre, The University of Sydney, Sydney, NSW, 2006, Australia
- Chronic Diseases Research Theme, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, 2006, Australia
| | - Markus J Hofer
- Charles Perkins Centre, The University of Sydney, Sydney, NSW, 2006, Australia
- School of Life and Environmental Sciences, The University of Sydney, Sydney, NSW, 2006, Australia
- The University of Sydney Institute for Infectious Diseases, The University of Sydney, Sydney, NSW, 2006, Australia
| | - Nicholas J C King
- Viral Immunopathology Laboratory, Infection, Immunity and Inflammation Research Theme, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, 2006, Australia.
- Sydney Cytometry, The University of Sydney and Centenary Institute, Sydney, NSW, 2006, Australia.
- Ramaciotti Facility for Human Systems Biology, The University of Sydney and Centenary Institute, Sydney, NSW, 2006, Australia.
- Charles Perkins Centre, The University of Sydney, Sydney, NSW, 2006, Australia.
- The University of Sydney Institute for Infectious Diseases, The University of Sydney, Sydney, NSW, 2006, Australia.
- Sydney Nano, The University of Sydney, Sydney, NSW, 2006, Australia.
| |
Collapse
|
8
|
Wishart CL, Spiteri AG, Locatelli G, King NJC. Integrating transcriptomic datasets across neurological disease identifies unique myeloid subpopulations driving disease-specific signatures. Glia 2023; 71:904-925. [PMID: 36527260 PMCID: PMC10952672 DOI: 10.1002/glia.24314] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 11/06/2022] [Accepted: 11/23/2022] [Indexed: 12/23/2022]
Abstract
Microglia and bone marrow-derived monocytes are key elements of central nervous system (CNS) inflammation, both capable of enhancing and dampening immune-mediated pathology. However, the study-specific focus on individual cell types, disease models or experimental approaches has limited our ability to infer common and disease-specific responses. This meta-analysis integrates bulk and single-cell transcriptomic datasets of microglia and monocytes from disease models of autoimmunity, neurodegeneration, sterile injury, and infection to build a comprehensive resource connecting myeloid responses across CNS disease. We demonstrate that the bulk microglial and monocyte program is highly contingent on the disease environment, challenging the notion of a universal microglial disease signature. Integration of six single-cell RNA-sequencing datasets revealed that these disease-specific signatures are likely driven by differing proportions of unique myeloid subpopulations that were individually expanded in different disease settings. These subsets were functionally-defined as neurodegeneration-associated, inflammatory, interferon-responsive, phagocytic, antigen-presenting, and lipopolysaccharide-responsive cellular states, revealing a core set of myeloid responses at the single-cell level that are conserved across CNS pathology. Showcasing the predictive and practical value of this resource, we performed differential expression analysis on microglia and monocytes across disease and identified Cd81 as a new neuroinflammatory-stable gene that accurately identified microglia and distinguished them from monocyte-derived cells across all experimental models at both the bulk and single-cell level. Together, this resource dissects the influence of disease environment on shared immune response programmes to build a unified perspective of myeloid behavior across CNS pathology.
Collapse
Affiliation(s)
- Claire L. Wishart
- Infection, Immunity, Inflammation Research Theme, School of Medical Sciences, Faculty of Medicine and HealthThe University of SydneySydneyNew South WalesAustralia
- Sydney Cytometry FacilityThe University of Sydney and Centenary InstituteSydneyNew South WalesAustralia
- Ramaciotti Facility for Human Systems BiologyThe University of Sydney and Centenary InstituteSydneyNew South WalesAustralia
- Charles Perkins CentreThe University of SydneySydneyNew South WalesAustralia
| | - Alanna G. Spiteri
- Infection, Immunity, Inflammation Research Theme, School of Medical Sciences, Faculty of Medicine and HealthThe University of SydneySydneyNew South WalesAustralia
- Sydney Cytometry FacilityThe University of Sydney and Centenary InstituteSydneyNew South WalesAustralia
- Ramaciotti Facility for Human Systems BiologyThe University of Sydney and Centenary InstituteSydneyNew South WalesAustralia
- Charles Perkins CentreThe University of SydneySydneyNew South WalesAustralia
| | - Giuseppe Locatelli
- Theodor Kocher InstituteUniversity of BernBernSwitzerland
- Novartis Institutes for BioMedical ResearchNovartisBaselSwitzerland
| | - Nicholas J. C. King
- Infection, Immunity, Inflammation Research Theme, School of Medical Sciences, Faculty of Medicine and HealthThe University of SydneySydneyNew South WalesAustralia
- Sydney Cytometry FacilityThe University of Sydney and Centenary InstituteSydneyNew South WalesAustralia
- Ramaciotti Facility for Human Systems BiologyThe University of Sydney and Centenary InstituteSydneyNew South WalesAustralia
- Charles Perkins CentreThe University of SydneySydneyNew South WalesAustralia
- Sydney Institute for Infectious Diseases, Faculty of Medicine and HealthThe University of SydneySydneyNew South WalesAustralia
- The University of Sydney Nano Institute, Faculty of ScienceThe University of SydneySydneyNew South WalesAustralia
| |
Collapse
|
9
|
RNA Editing Alterations Define Disease Manifestations in the Progression of Experimental Autoimmune Encephalomyelitis (EAE). Cells 2022; 11:cells11223582. [PMID: 36429012 PMCID: PMC9688714 DOI: 10.3390/cells11223582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 11/01/2022] [Accepted: 11/09/2022] [Indexed: 11/16/2022] Open
Abstract
RNA editing is an epitranscriptomic modification, leading to targeted changes in RNA transcripts. It is mediated by the action of ADAR (adenosine deaminases acting on double-stranded (ds) RNA and APOBEC (apolipoprotein B mRNA editing enzyme catalytic polypeptide-like) deaminases and appears to play a major role in the pathogenesis of many diseases. Here, we assessed its role in experimental autoimmune encephalomyelitis (EAE), a widely used non-clinical model of autoimmune inflammatory diseases of the central nervous system (CNS), which resembles many aspects of human multiple sclerosis (MS). We have analyzed in silico data from microglia isolated at different timepoints through disease progression to identify the global editing events and validated the selected targets in murine tissue samples. To further evaluate the functional role of RNA editing, we induced EAE in transgenic animals lacking expression of APOBEC-1. We found that RNA-editing events, mediated by the APOBEC and ADAR deaminases, are significantly reduced throughout the course of disease, possibly affecting the protein expression necessary for normal neurological function. Moreover, the severity of the EAE model was significantly higher in APOBEC-1 knock-out mice, compared to wild-type controls. Our results implicate regulatory epitranscriptomic mechanisms in EAE pathogenesis that could be extrapolated to MS and other neurodegenerative disorders (NDs) with common clinical and molecular features.
Collapse
|
10
|
Mason HD, McGavern DB. How the immune system shapes neurodegenerative diseases. Trends Neurosci 2022; 45:733-748. [PMID: 36075783 PMCID: PMC9746609 DOI: 10.1016/j.tins.2022.08.001] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 06/10/2022] [Accepted: 08/01/2022] [Indexed: 12/15/2022]
Abstract
Neurodegenerative diseases are a major cause of death and disability worldwide and are influenced by many factors including age, genetics, and injuries. While these diseases are often thought to result from the accumulation and spread of aberrant proteins, recent studies have demonstrated that they can be shaped by the innate and adaptive immune system. Resident myeloid cells typically mount a sustained response to the degenerating CNS, but peripheral leukocytes such as T and B cells can also alter disease trajectories. Here, we review the sometimes-dichotomous roles played by immune cells during neurodegenerative diseases and explore how brain trauma can serve as a disease initiator or accelerant. We also offer insights into how failure to properly resolve a CNS injury might promote the development of a neurodegenerative disease.
Collapse
Affiliation(s)
- Hannah D Mason
- Viral Immunology and Intravital Imaging Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Dorian B McGavern
- Viral Immunology and Intravital Imaging Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
11
|
Biswas DD, Martin RK, Brown LN, Mockenhaupt K, Gupta AS, Surace MJ, Tharakan A, Yester JW, Bhardwaj R, Conrad DH, Kordula T. Cellular inhibitor of apoptosis 2 (cIAP2) restricts neuroinflammation during experimental autoimmune encephalomyelitis. J Neuroinflammation 2022; 19:158. [PMID: 35718775 PMCID: PMC9208101 DOI: 10.1186/s12974-022-02527-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Accepted: 06/11/2022] [Indexed: 11/10/2022] Open
Abstract
Background Immune activation, neuroinflammation, and cell death are the hallmarks of multiple sclerosis (MS), which is an autoimmune demyelinating disease of the central nervous system (CNS). It is well-documented that the cellular inhibitor of apoptosis 2 (cIAP2) is induced by inflammatory stimuli and regulates adaptive and innate immune responses, cell death, and the production of inflammatory mediators. However, the impact of cIAP2 on neuroinflammation associated with MS and disease severity remains unknown.
Methods We used experimental autoimmune encephalomyelitis (EAE), a widely used mouse model of MS, to assess the effect of cIAP2 deletion on disease outcomes. We performed a detailed analysis on the histological, cellular, and molecular levels. We generated and examined bone-marrow chimeras to identify the cIAP2-deficient cells that are critical to the disease outcomes. Results cIAP2−/− mice exhibited increased EAE severity, increased CD4+ T cell infiltration, enhanced proinflammatory cytokine/chemokine expression, and augmented demyelination. This phenotype was driven by cIAP2-deficient non-hematopoietic cells. cIAP2 protected oligodendrocytes from cell death during EAE by limiting proliferation and activation of brain microglia. This protective role was likely exerted by cIAP2-mediated inhibition of the non-canonical NLRP3/caspase-8-dependent myeloid cell activation during EAE. Conclusions Our findings suggest that cIAP2 is needed to modulate neuroinflammation, cell death, and survival during EAE. Significantly, our data demonstrate the critical role of cIAP2 in limiting the activation of microglia during EAE, which could be explored for developing MS therapeutics in the future. Supplementary Information The online version contains supplementary material available at 10.1186/s12974-022-02527-6.
Collapse
Affiliation(s)
- Debolina D Biswas
- Department of Biochemistry and Molecular Biology, School of Medicine and the Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, 23298, USA
| | - Rebecca K Martin
- Department of Microbiology and Immunology, School of Medicine and the Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, 23298, USA
| | - LaShardai N Brown
- Department of Biochemistry and Molecular Biology, School of Medicine and the Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, 23298, USA
| | - Karli Mockenhaupt
- Department of Biochemistry and Molecular Biology, School of Medicine and the Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, 23298, USA
| | - Angela S Gupta
- Department of Biochemistry and Molecular Biology, School of Medicine and the Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, 23298, USA
| | - Michael J Surace
- Department of Biochemistry and Molecular Biology, School of Medicine and the Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, 23298, USA
| | - Anuj Tharakan
- Department of Biochemistry and Molecular Biology, School of Medicine and the Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, 23298, USA
| | - Jessie W Yester
- Department of Biochemistry and Molecular Biology, School of Medicine and the Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, 23298, USA
| | - Reetika Bhardwaj
- Department of Biochemistry and Molecular Biology, School of Medicine and the Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, 23298, USA
| | - Daniel H Conrad
- Department of Microbiology and Immunology, School of Medicine and the Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, 23298, USA
| | - Tomasz Kordula
- Department of Biochemistry and Molecular Biology, School of Medicine and the Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, 23298, USA.
| |
Collapse
|
12
|
Spiteri AG, Wishart CL, Pamphlett R, Locatelli G, King NJC. Microglia and monocytes in inflammatory CNS disease: integrating phenotype and function. Acta Neuropathol 2022; 143:179-224. [PMID: 34853891 PMCID: PMC8742818 DOI: 10.1007/s00401-021-02384-2] [Citation(s) in RCA: 114] [Impact Index Per Article: 38.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 11/11/2021] [Accepted: 11/11/2021] [Indexed: 02/08/2023]
Abstract
In neurological diseases, the actions of microglia, the resident myeloid cells of the CNS parenchyma, may diverge from, or intersect with, those of recruited monocytes to drive immune-mediated pathology. However, defining the precise roles of each cell type has historically been impeded by the lack of discriminating markers and experimental systems capable of accurately identifying them. Our ability to distinguish microglia from monocytes in neuroinflammation has advanced with single-cell technologies, new markers and drugs that identify and deplete them, respectively. Nevertheless, the focus of individual studies on particular cell types, diseases or experimental approaches has limited our ability to connect phenotype and function more widely and across diverse CNS pathologies. Here, we critically review, tabulate and integrate the disease-specific functions and immune profiles of microglia and monocytes to provide a comprehensive atlas of myeloid responses in viral encephalitis, demyelination, neurodegeneration and ischemic injury. In emphasizing the differential roles of microglia and monocytes in the severe neuroinflammatory disease of viral encephalitis, we connect inflammatory pathways common to equally incapacitating diseases with less severe inflammation. We examine these findings in the context of human studies and highlight the benefits and inherent limitations of animal models that may impede or facilitate clinical translation. This enables us to highlight common and contrasting, non-redundant and often opposing roles of microglia and monocytes in disease that could be targeted therapeutically.
Collapse
|
13
|
Zelenka L, Pägelow D, Krüger C, Seele J, Ebner F, Rausch S, Rohde M, Lehnardt S, van Vorst K, Fulde M. Novel protocol for the isolation of highly purified neonatal murine microglia and astrocytes. J Neurosci Methods 2022; 366:109420. [PMID: 34808220 DOI: 10.1016/j.jneumeth.2021.109420] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 11/02/2021] [Accepted: 11/11/2021] [Indexed: 01/03/2023]
Abstract
BACKGROUND The crosstalk and reactivity of the cell type glia, especially microglia and astrocytes, have progressively gathered research attention in understanding proper brain function regulated by the innate immune response. Therefore, methods to isolate highly viable and pure glia for the analysis on a cell-specific level are indispensable. NEW METHOD We modified previously established techniques: Animal numbers were reduced by multiple microglial harvests from the same mixed glial culture, thereby maximizing microglial yields following the principles of the 3Rs (replacement, reduction, and refinement). We optimized Magnetic-activated cell sorting (MACS®) of microglia and astrocytes by applying cultivated primary glial cell suspensions instead of directly sorting dissociated single cell suspension. RESULTS We generated highly viable and pure microglia and astrocytes derived from a single mixed culture with a purity of ~99%, as confirmed by FACS analysis. Field emission scanning electron microscopy (FESEM) demonstrated integrity of the MACS-purified glial cells. Tumor necrosis factor (TNF) and Interleukin-10 (IL-10) ELISA confirmed pro- and anti-inflammatory responses to be functional in purified glia, but significantly weakened compared to non-purified cells, further highlighting the importance of cellular crosstalk for proper immune activation. COMPARISON WITH EXISTING METHOD(S) Unlike previous studies that either isolated a single type of glia or displayed a substantial proportion of contamination with other cell types, we achieved isolation of both microglia and astrocytes at an increased purity (99-100%). CONCLUSIONS We have created an optimized protocol for the efficient purification of both primary microglia and astrocytes. Our results clearly demonstrate the importance of purity in glial cell cultivation in order to examine immune responses, which particularly holds true for astrocytes. We propose the novel protocol as a tool to investigate the cell type-specific crosstalk between microglia and astrocytes in the frame of CNS diseases.
Collapse
Affiliation(s)
- Laura Zelenka
- Institute of Microbiology and Epizootics, Centre of Infection Medicine, Freie Universität Berlin, Robert-von-Ostertag-Straße 7-13, 14163 Berlin, Germany
| | - Dennis Pägelow
- Institute of Microbiology and Epizootics, Centre of Infection Medicine, Freie Universität Berlin, Robert-von-Ostertag-Straße 7-13, 14163 Berlin, Germany
| | - Christina Krüger
- Institute of Cell Biology and Neurobiology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Jana Seele
- University Medical Center Göttingen, Institute of Neuropathology, Göttingen, Germany
| | - Friederike Ebner
- Freie Universität Berlin, Institute of Immunology, Berlin, Germany
| | - Sebastian Rausch
- Freie Universität Berlin, Institute of Immunology, Berlin, Germany
| | - Manfred Rohde
- Central Facility for Microscopy, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Seija Lehnardt
- Institute of Cell Biology and Neurobiology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany; Department of Neurology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Kira van Vorst
- Institute of Microbiology and Epizootics, Centre of Infection Medicine, Freie Universität Berlin, Robert-von-Ostertag-Straße 7-13, 14163 Berlin, Germany
| | - Marcus Fulde
- Institute of Microbiology and Epizootics, Centre of Infection Medicine, Freie Universität Berlin, Robert-von-Ostertag-Straße 7-13, 14163 Berlin, Germany.
| |
Collapse
|
14
|
Park J, Choi SW, Cha BG, Kim J, Kang SJ. Alternative Activation of Macrophages through Interleukin-13-Loaded Extra-Large-Pore Mesoporous Silica Nanoparticles Suppresses Experimental Autoimmune Encephalomyelitis. ACS Biomater Sci Eng 2021; 7:4446-4453. [PMID: 34435775 DOI: 10.1021/acsbiomaterials.1c00946] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Multiple sclerosis (MS) treatment via cytokine-mediated immunomodulation has been hampered by the difficulty with which cytokines can be stably and noninvasively delivered to the central nervous system. Here, we show that interleukin (IL)-13 packaged in extra-large-pore mesoporous silica nanoparticles (XL-MSNs) is protected from degradation and directs the alternative activation of macrophages both in vitro and in vivo. Furthermore, the noninvasive intranasal delivery of IL-13-loaded XL-MSNs ameliorated the symptoms of experimental autoimmune encephalomyelitis, a murine model of MS, accompanied by the induction of chemokines orchestrating immune cell infiltration. These results demonstrate the therapeutic potential of IL-13-loaded XL-MSNs for MS patients.
Collapse
Affiliation(s)
- Jiyeon Park
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea
| | - Seung Woo Choi
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences & Technology (SAIHST), Sungkyunkwan University (SKKU), Seoul 06355, Republic of Korea
| | - Bong Geun Cha
- School of Chemical Engineering, Sungkyunkwan University (SKKU), Suwon 16419, Republic of Korea
| | - Jaeyun Kim
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences & Technology (SAIHST), Sungkyunkwan University (SKKU), Seoul 06355, Republic of Korea.,School of Chemical Engineering, Sungkyunkwan University (SKKU), Suwon 16419, Republic of Korea.,Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University (SKKU), Suwon 16419, Republic of Korea.,Institute of Quantum Biophysics (IQB), Sungkyunkwan University (SKKU), Suwon 16419, Republic of Korea
| | - Suk-Jo Kang
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea
| |
Collapse
|
15
|
Radandish M, Khalilian P, Esmaeil N. The Role of Distinct Subsets of Macrophages in the Pathogenesis of MS and the Impact of Different Therapeutic Agents on These Populations. Front Immunol 2021; 12:667705. [PMID: 34489926 PMCID: PMC8417824 DOI: 10.3389/fimmu.2021.667705] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Accepted: 07/31/2021] [Indexed: 01/03/2023] Open
Abstract
Multiple sclerosis (MS) is a demyelinating inflammatory disorder of the central nervous system (CNS). Besides the vital role of T cells, other immune cells, including B cells, innate immune cells, and macrophages (MФs), also play a critical role in MS pathogenesis. Tissue-resident MФs in the brain’s parenchyma, known as microglia and monocyte-derived MФs, enter into the CNS following alterations in CNS homeostasis that induce inflammatory responses in MS. Although the neuroprotective and anti-inflammatory actions of monocyte-derived MФs and resident MФs are required to maintain CNS tolerance, they can release inflammatory cytokines and reactivate primed T cells during neuroinflammation. In the CNS of MS patients, elevated myeloid cells and activated MФs have been found and associated with demyelination and axonal loss. Thus, according to the role of MФs in neuroinflammation, they have attracted attention as a therapeutic target. Also, due to their different origin, location, and turnover, other strategies may require to target the various myeloid cell populations. Here we review the role of distinct subsets of MФs in the pathogenesis of MS and different therapeutic agents that target these cells.
Collapse
Affiliation(s)
- Maedeh Radandish
- Department of Immunology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Parvin Khalilian
- Department of Immunology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Nafiseh Esmaeil
- Department of Immunology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran.,Environment Research Center, Research Institute for Primordial Prevention of Non-Communicable Disease, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
16
|
Salcman B, Affleck K, Bulfone-Paus S. P2X Receptor-Dependent Modulation of Mast Cell and Glial Cell Activities in Neuroinflammation. Cells 2021; 10:cells10092282. [PMID: 34571930 PMCID: PMC8471135 DOI: 10.3390/cells10092282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 08/23/2021] [Accepted: 08/30/2021] [Indexed: 11/27/2022] Open
Abstract
Localisation of mast cells (MCs) at the abluminal side of blood vessels in the brain favours their interaction with glial cells, neurons, and endothelial cells, resulting in the activation of these cells and the release of pro-inflammatory mediators. In turn, stimulation of glial cells, such as microglia, astrocytes, and oligodendrocytes may result in the modulation of MC activities. MCs, microglia, astrocytes, and oligodendrocytes all express P2X receptors (P2XRs) family members that are selectively engaged by ATP. As increased concentrations of extracellular adenosine 5′-triphosphate (ATP) are present in the brain in neuropathological conditions, P2XR activation in MCs and glial cells contributes to the control of their communication and amplification of the inflammatory response. In this review we discuss P2XR-mediated MC activation, its bi-directional effect on microglia, astrocytes and oligodendrocytes and role in neuroinflammation.
Collapse
Affiliation(s)
- Barbora Salcman
- Lydia Becker Institute of Immunology and Inflammation, Manchester Collaborative Centre for Inflammation Research, University of Manchester, Manchester M13 9NT, UK;
| | - Karen Affleck
- GlaxoSmithKline, Immunology Research Unit, Stevenage SG1 2NY, UK;
| | - Silvia Bulfone-Paus
- Lydia Becker Institute of Immunology and Inflammation, Manchester Collaborative Centre for Inflammation Research, University of Manchester, Manchester M13 9NT, UK;
- Correspondence:
| |
Collapse
|
17
|
Mitchell D, Shireman J, Sierra Potchanant EA, Lara-Velazquez M, Dey M. Neuroinflammation in Autoimmune Disease and Primary Brain Tumors: The Quest for Striking the Right Balance. Front Cell Neurosci 2021; 15:716947. [PMID: 34483843 PMCID: PMC8414998 DOI: 10.3389/fncel.2021.716947] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Accepted: 07/26/2021] [Indexed: 12/25/2022] Open
Abstract
According to classical dogma, the central nervous system (CNS) is defined as an immune privileged space. The basis of this theory was rooted in an incomplete understanding of the CNS microenvironment, however, recent advances such as the identification of resident dendritic cells (DC) in the brain and the presence of CNS lymphatics have deepened our understanding of the neuro-immune axis and revolutionized the field of neuroimmunology. It is now understood that many pathological conditions induce an immune response in the CNS, and that in many ways, the CNS is an immunologically distinct organ. Hyperactivity of neuro-immune axis can lead to primary neuroinflammatory diseases such as multiple sclerosis and antibody-mediated encephalitis, whereas immunosuppressive mechanisms promote the development and survival of primary brain tumors. On the therapeutic front, attempts are being made to target CNS pathologies using various forms of immunotherapy. One of the most actively investigated areas of CNS immunotherapy is for the treatment of glioblastoma (GBM), the most common primary brain tumor in adults. In this review, we provide an up to date overview of the neuro-immune axis in steady state and discuss the mechanisms underlying neuroinflammation in autoimmune neuroinflammatory disease as well as in the development and progression of brain tumors. In addition, we detail the current understanding of the interactions that characterize the primary brain tumor microenvironment and the implications of the neuro-immune axis on the development of successful therapeutic strategies for the treatment of CNS malignancies.
Collapse
Affiliation(s)
- Dana Mitchell
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Jack Shireman
- Dey Malignant Brain Tumor Laboratory, Department of Neurological Surgery, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States
| | | | - Montserrat Lara-Velazquez
- Dey Malignant Brain Tumor Laboratory, Department of Neurological Surgery, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States
| | - Mahua Dey
- Dey Malignant Brain Tumor Laboratory, Department of Neurological Surgery, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States
| |
Collapse
|
18
|
Goldfarb S, Fainstein N, Ganz T, Vershkov D, Lachish M, Ben-Hur T. Electric neurostimulation regulates microglial activation via retinoic acid receptor α signaling. Brain Behav Immun 2021; 96:40-53. [PMID: 33989746 DOI: 10.1016/j.bbi.2021.05.007] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 04/20/2021] [Accepted: 05/09/2021] [Indexed: 12/14/2022] Open
Abstract
Brain stimulation by electroconvulsive therapy is effective in neuropsychiatric disorders by unknown mechanisms. Microglial toxicity plays key role in neuropsychiatric, neuroinflammatory and degenerative diseases. We examined the mechanism by which electroconvulsive seizures (ECS) regulates microglial phenotype and response to stimuli. Microglial responses were examined by morphological analysis, Iba1 and cytokine expression. ECS did not affect resting microglial phenotype or morphology but regulated their activation by Lipopolysaccharide stimulation. Microglia were isolated after ECS or sham sessions in naïve mice for transcriptome analysis. RNA sequencing identified 141 differentially expressed genes. ECS modulated multiple immune-associated gene families and attenuated neurotoxicity-associated gene expression. Blood brain barrier was examined by injecting Biocytin-TMR tracer. There was no breakdown of the BBB, nor increase in gene-signature of peripheral monocytes, suggesting that ECS effect is mainly on resident microglia. Unbiased analysis of regulatory sequences identified the induction of microglial retinoic acid receptor α (RARα) gene expression and a putative common RARα-binding motif in multiple ECS-upregulated genes. The effects of AM580, a selective RARα agonist on microglial response to LPS was examined in vitro. AM580 prevented LPS-induced cytokine expression and reactive oxygen species production. Chronic murine experimental autoimmune encephalomyelitis (EAE) was utilized to confirm the role RARα signaling as mediator of ECS-induced transcriptional pathway in regulating microglial toxicity. Continuous intracerebroventricular delivery of AM580 attenuated effectively EAE severity. In conclusion, ECS regulates CNS innate immune system responses by activating microglial retinoic acid receptor α pathway, signifying a novel therapeutic approach for chronic neuroinflammatory, neuropsychiatric and neurodegenerative diseases.
Collapse
Affiliation(s)
- Smadar Goldfarb
- Faculty of Medicine, Hebrew University of Jerusalem, Israel; The Department of Neurology, The Agnes Ginges Center for Human Neurogenetics, Hadassah - Hebrew University Medical Center, Jerusalem, Israel
| | - Nina Fainstein
- Faculty of Medicine, Hebrew University of Jerusalem, Israel; The Department of Neurology, The Agnes Ginges Center for Human Neurogenetics, Hadassah - Hebrew University Medical Center, Jerusalem, Israel
| | - Tal Ganz
- Faculty of Medicine, Hebrew University of Jerusalem, Israel
| | - Dan Vershkov
- Faculty of Medicine, Hebrew University of Jerusalem, Israel; The Department of Neurology, The Agnes Ginges Center for Human Neurogenetics, Hadassah - Hebrew University Medical Center, Jerusalem, Israel; The Azrieli Center for Stem Cells and Genetic Research, Department of Genetics, Silberman Institute of Life Sciences, The Hebrew University, Jerusalem, Israel
| | - Marva Lachish
- Faculty of Medicine, Hebrew University of Jerusalem, Israel; The Department of Neurology, The Agnes Ginges Center for Human Neurogenetics, Hadassah - Hebrew University Medical Center, Jerusalem, Israel
| | - Tamir Ben-Hur
- Faculty of Medicine, Hebrew University of Jerusalem, Israel; The Department of Neurology, The Agnes Ginges Center for Human Neurogenetics, Hadassah - Hebrew University Medical Center, Jerusalem, Israel.
| |
Collapse
|
19
|
Spiteri AG, Terry RL, Wishart CL, Ashhurst TM, Campbell IL, Hofer MJ, King NJC. High-parameter cytometry unmasks microglial cell spatio-temporal response kinetics in severe neuroinflammatory disease. J Neuroinflammation 2021; 18:166. [PMID: 34311763 PMCID: PMC8314570 DOI: 10.1186/s12974-021-02214-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Accepted: 07/07/2021] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Differentiating infiltrating myeloid cells from resident microglia in neuroinflammatory disease is challenging, because bone marrow-derived inflammatory monocytes infiltrating the inflamed brain adopt a 'microglia-like' phenotype. This precludes the accurate identification of either cell type without genetic manipulation, which is important to understand their temporal contribution to disease and inform effective intervention in its pathogenesis. During West Nile virus (WNV) encephalitis, widespread neuronal infection drives substantial CNS infiltration of inflammatory monocytes, causing severe immunopathology and/or death, but the role of microglia in this remains unclear. METHODS Using high-parameter cytometry and dimensionality-reduction, we devised a simple, novel gating strategy to identify microglia and infiltrating myeloid cells during WNV-infection. Validating our strategy, we (1) blocked the entry of infiltrating myeloid populations from peripheral blood using monoclonal blocking antibodies, (2) adoptively transferred BM-derived monocytes and tracked their phenotypic changes after infiltration and (3) labelled peripheral leukocytes that infiltrate into the brain with an intravenous dye. We demonstrated that myeloid immigrants populated only the identified macrophage gates, while PLX5622 depletion reduced all 4 subsets defined by the microglial gates. RESULTS Using this gating approach, we identified four consistent microglia subsets in the homeostatic and WNV-infected brain. These were P2RY12hi CD86-, P2RY12hi CD86+ and P2RY12lo CD86- P2RY12lo CD86+. During infection, 2 further populations were identified as 'inflammatory' and 'microglia-like' macrophages, recruited from the bone marrow. Detailed kinetic analysis showed significant increases in the proportions of both P2RY12lo microglia subsets in all anatomical areas, largely at the expense of the P2RY12hi CD86- subset, with the latter undergoing compensatory proliferation, suggesting replenishment of, and differentiation from this subset in response to infection. Microglia altered their morphology early in infection, with all cells adopting temporal and regional disease-specific phenotypes. Late in disease, microglia produced IL-12, downregulated CX3CR1, F4/80 and TMEM119 and underwent apoptosis. Infiltrating macrophages expressed both TMEM119 and P2RY12 de novo, with the microglia-like subset notably exhibiting the highest proportional myeloid population death. CONCLUSIONS Our approach enables detailed kinetic analysis of resident vs infiltrating myeloid cells in a wide range of neuroinflammatory models without non-physiological manipulation. This will more clearly inform potential therapeutic approaches that specifically modulate these cells.
Collapse
Affiliation(s)
- Alanna G Spiteri
- Discipline of Pathology, Faculty of Medicine and Health, School of Medical Sciences, The University of Sydney, Sydney, Australia
- Charles Perkins Centre, The University of Sydney, Sydney, Australia
| | - Rachel L Terry
- Discipline of Pathology, Faculty of Medicine and Health, School of Medical Sciences, The University of Sydney, Sydney, Australia
- Charles Perkins Centre, The University of Sydney, Sydney, Australia
- Current Address: Children's Cancer Institute, Randwick, New South Wales, Australia
- Current Affiliation: Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Claire L Wishart
- Discipline of Pathology, Faculty of Medicine and Health, School of Medical Sciences, The University of Sydney, Sydney, Australia
- Charles Perkins Centre, The University of Sydney, Sydney, Australia
| | - Thomas M Ashhurst
- Discipline of Pathology, Faculty of Medicine and Health, School of Medical Sciences, The University of Sydney, Sydney, Australia
- Charles Perkins Centre, The University of Sydney, Sydney, Australia
- Sydney Cytometry Facility, The University of Sydney and Centenary Institute, Sydney, Australia
- Ramaciotti Facility for Human Systems Biology, The University of Sydney and Centenary Institute, Sydney, Australia
- Marie Bashir Institute for Infectious Diseases and Biosecurity (MBI), Faculty of Medicine and Health, Sydney Medical School, The University of Sydney, Sydney, Australia
| | - Iain L Campbell
- Marie Bashir Institute for Infectious Diseases and Biosecurity (MBI), Faculty of Medicine and Health, Sydney Medical School, The University of Sydney, Sydney, Australia
- School of Life and Environmental Sciences, The University of Sydney, Sydney, Australia
| | - Markus J Hofer
- Charles Perkins Centre, The University of Sydney, Sydney, Australia
- Marie Bashir Institute for Infectious Diseases and Biosecurity (MBI), Faculty of Medicine and Health, Sydney Medical School, The University of Sydney, Sydney, Australia
- School of Life and Environmental Sciences, The University of Sydney, Sydney, Australia
| | - Nicholas J C King
- Discipline of Pathology, Faculty of Medicine and Health, School of Medical Sciences, The University of Sydney, Sydney, Australia.
- Charles Perkins Centre, The University of Sydney, Sydney, Australia.
- Sydney Cytometry Facility, The University of Sydney and Centenary Institute, Sydney, Australia.
- Ramaciotti Facility for Human Systems Biology, The University of Sydney and Centenary Institute, Sydney, Australia.
- Marie Bashir Institute for Infectious Diseases and Biosecurity (MBI), Faculty of Medicine and Health, Sydney Medical School, The University of Sydney, Sydney, Australia.
- Nano Institute, The University of Sydney, Sydney, Australia.
| |
Collapse
|
20
|
Acharjee S, Gordon PMK, Lee BH, Read J, Workentine ML, Sharkey KA, Pittman QJ. Characterization of microglial transcriptomes in the brain and spinal cord of mice in early and late experimental autoimmune encephalomyelitis using a RiboTag strategy. Sci Rep 2021; 11:14319. [PMID: 34253764 PMCID: PMC8275680 DOI: 10.1038/s41598-021-93590-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Accepted: 06/25/2021] [Indexed: 12/29/2022] Open
Abstract
Microglia play an important role in the pathogenesis of multiple sclerosis and the mouse model of MS, experimental autoimmune encephalomyelitis (EAE). To more fully understand the role of microglia in EAE we characterized microglial transcriptomes before the onset of motor symptoms (pre-onset) and during symptomatic EAE. We compared the transcriptome in brain, where behavioral changes are initiated, and spinal cord, where damage is revealed as motor and sensory deficits. We used a RiboTag strategy to characterize ribosome-bound mRNA only in microglia without incurring possible transcriptional changes after cell isolation. Brain and spinal cord samples clustered separately at both stages of EAE, indicating regional heterogeneity. Differences in gene expression were observed in the brain and spinal cord of pre-onset and symptomatic animals with most profound effects in the spinal cord of symptomatic animals. Canonical pathway analysis revealed changes in neuroinflammatory pathways, immune functions and enhanced cell division in both pre-onset and symptomatic brain and spinal cord. We also observed a continuum of many pathways at pre-onset stage that continue into the symptomatic stage of EAE. Our results provide additional evidence of regional and temporal heterogeneity in microglial gene expression patterns that may help in understanding mechanisms underlying various symptomology in MS.
Collapse
Affiliation(s)
- Shaona Acharjee
- Hotchkiss Brain Institute, Snyder Institute for Chronic Diseases, Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, 3330 Hospital Drive NW, Calgary, AB, T2N 4N1, Canada
| | - Paul M K Gordon
- Centre for Health Genomics and Informatics, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Benjamin H Lee
- Hotchkiss Brain Institute, Snyder Institute for Chronic Diseases, Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, 3330 Hospital Drive NW, Calgary, AB, T2N 4N1, Canada
| | - Justin Read
- Hotchkiss Brain Institute, Snyder Institute for Chronic Diseases, Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, 3330 Hospital Drive NW, Calgary, AB, T2N 4N1, Canada
| | - Matthew L Workentine
- Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, Canada
| | - Keith A Sharkey
- Hotchkiss Brain Institute, Snyder Institute for Chronic Diseases, Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, 3330 Hospital Drive NW, Calgary, AB, T2N 4N1, Canada
| | - Quentin J Pittman
- Hotchkiss Brain Institute, Snyder Institute for Chronic Diseases, Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, 3330 Hospital Drive NW, Calgary, AB, T2N 4N1, Canada.
| |
Collapse
|
21
|
Li Q, Wen S, Ye W, Zhao S, Liu X. The potential roles of m 6A modification in regulating the inflammatory response in microglia. J Neuroinflammation 2021; 18:149. [PMID: 34225746 PMCID: PMC8259013 DOI: 10.1186/s12974-021-02205-z] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Accepted: 06/23/2021] [Indexed: 01/15/2023] Open
Abstract
BACKGROUND Microglia are key regulators of the inflammatory response in the brain. Adenosine in RNAs can be converted to m6A (N6-methyladenosine), which regulates RNA metabolism and functions as a key epitranscriptomic modification. The m6A modification pattern and m6A-related signatures under pro-inflammatory and anti-inflammatory conditions of microglia remain unclear. METHODS Primary rat microglia were differentiated into pro-inflammatory M1-like (M1-L), anti-inflammatory M2-like (M2-L), and resting, unstimulated (M0-L) phenotypes. m6A mRNA and lncRNA epitranscriptomic microarray analyses were performed, and pathway analysis was conducted to understand the functional implications of m6A methylation in mRNAs and lncRNAs. The m6A methylation level and gene expression of mRNAs and lncRNAs were subsequently verified by m6A Me-RIP and qRT-PCR. RESULTS A total of 1588 mRNAs and 340 lncRNAs, 315 mRNAs and 38 lncRNAs, and 521 mRNAs and 244 lncRNAs were differentially m6A methylated between M1-L and M0-L (M1-L/M0-L), M2-L and M0-L (M2-L/M0-L), M2-L and M1-L (M2-L/M1-L), respectively. Furthermore, 4902 mRNAs, 4676 mRNAs, and 5095 mRNAs were identified distinctively expressed in M1-L/M0-L, M2-L/M0-L, and M2-L/M1-L, respectively. Pathway analysis of differentially m6A methylated mRNAs and lncRNAs in M1-L/M0-L identified immune system, signal transduction, and protein degradation processes. In contrast, the distinct m6A methylated mRNAs in M2-L/M0-L were involved in genetic information processing, metabolism, cellular processes, and neurodegenerative disease-related pathways. We validated m6A methylation and the expression levels of five mRNAs and five lncRNAs, which were involved in upregulated pathways in M1-L/M0-L, and five mRNAs involved in upregulated pathways in M2-L/M0-L. CONCLUSIONS These findings identify a distinct m6A epitranscriptome in microglia, and which may serve as novel and useful regulator during pro-inflammatory and anti-inflammatory response of microglia.
Collapse
Affiliation(s)
- Qi Li
- China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, 119 South Fourth Ring West Road, Fengtai District, Beijing, 100070, People's Republic of China
| | - Shaohong Wen
- China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, 119 South Fourth Ring West Road, Fengtai District, Beijing, 100070, People's Republic of China
| | - Weizhen Ye
- China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, 119 South Fourth Ring West Road, Fengtai District, Beijing, 100070, People's Republic of China
| | - Shunying Zhao
- China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, 119 South Fourth Ring West Road, Fengtai District, Beijing, 100070, People's Republic of China
| | - Xiangrong Liu
- China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, 119 South Fourth Ring West Road, Fengtai District, Beijing, 100070, People's Republic of China.
| |
Collapse
|
22
|
Snijders GJLJ, Sneeboer MAM, Fernández-Andreu A, Udine E, Boks MP, Ormel PR, van Berlekom AB, van Mierlo HC, Bӧttcher C, Priller J, Raj T, Hol EM, Kahn RS, de Witte LD. Distinct non-inflammatory signature of microglia in post-mortem brain tissue of patients with major depressive disorder. Mol Psychiatry 2021; 26:3336-3349. [PMID: 33028963 DOI: 10.1038/s41380-020-00896-z] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 08/22/2020] [Accepted: 09/17/2020] [Indexed: 02/06/2023]
Abstract
Findings from epidemiological studies, biomarker measurements and animal experiments suggest a role for aberrant immune processes in the pathogenesis of major depressive disorder (MDD). Microglia, the resident immune cells of the brain, are likely to play a key role in these processes. Previous post-mortem studies reported conflicting findings regarding microglial activation and an in-depth profiling of those cells in MDD is lacking. The aim of this study was therefore to characterize the phenotype and function of microglia in MDD. We isolated microglia from post-mortem brain tissue of patients with MDD (n = 13-19) and control donors (n = 12-25). Using flow cytometry and quantitative Polymerase Chain Reaction (qPCR), we measured protein and mRNA levels of a panel of microglial markers across four different brain regions (medial frontal gyrus, superior temporal gyrus, thalamus, and subventricular zone). In MDD cases, we found a significant upregulation of CX3CR1 and TMEM119 mRNA expression and a downregulation of CD163 mRNA expression and CD14 protein expression across the four brain regions. Expression levels of microglial activation markers, such as HLA-DRA, IL6, and IL1β, as well as the inflammatory responses to lipopolysaccharide and dexamethasone were unchanged. Our findings suggest that microglia enhance homeostatic functions in MDD but are not immune activated.
Collapse
Affiliation(s)
- Gijsje J L J Snijders
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA. .,Department of Psychiatry, University Medical Center Utrecht Brain Center, Utrecht University, 3584, CG, Utrecht, The Netherlands.
| | - Marjolein A M Sneeboer
- Department of Psychiatry, University Medical Center Utrecht Brain Center, Utrecht University, 3584, CG, Utrecht, The Netherlands.,Department of Translational Neuroscience, University Medical Center Utrecht Brain Center, Utrecht University, 3584, CG, Utrecht, The Netherlands
| | - Alba Fernández-Andreu
- Department of Translational Neuroscience, University Medical Center Utrecht Brain Center, Utrecht University, 3584, CG, Utrecht, The Netherlands
| | - Evan Udine
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | - Marco P Boks
- Department of Psychiatry, University Medical Center Utrecht Brain Center, Utrecht University, 3584, CG, Utrecht, The Netherlands
| | - Paul R Ormel
- Department of Psychiatry, University Medical Center Utrecht Brain Center, Utrecht University, 3584, CG, Utrecht, The Netherlands.,Department of Translational Neuroscience, University Medical Center Utrecht Brain Center, Utrecht University, 3584, CG, Utrecht, The Netherlands
| | - Amber Berdenis van Berlekom
- Department of Psychiatry, University Medical Center Utrecht Brain Center, Utrecht University, 3584, CG, Utrecht, The Netherlands.,Department of Translational Neuroscience, University Medical Center Utrecht Brain Center, Utrecht University, 3584, CG, Utrecht, The Netherlands
| | - Hans C van Mierlo
- Department of Psychiatry, St. Antonius Hospital, Nieuwegein, Koekoekslaan 1, 3430, EM, Nieuwegein, The Netherlands
| | - Chotima Bӧttcher
- Department of Neuropsychiatry and Laboratory of Molecular Psychiatry, Charité-Universitätsmedizin Berlin, 10117, Berlin, Germany
| | - Josef Priller
- Department of Neuropsychiatry and Laboratory of Molecular Psychiatry, Charité-Universitätsmedizin Berlin, 10117, Berlin, Germany.,DZNE and BIH, 10117, Berlin, Germany.,University of Edinburgh and UK DRI, Edinburgh, EH16 4SB, UK
| | - Towfique Raj
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Elly M Hol
- Department of Translational Neuroscience, University Medical Center Utrecht Brain Center, Utrecht University, 3584, CG, Utrecht, The Netherlands.,Neuroimmunology, Netherlands Institute for Neuroscience, an institute of the royal academy of arts and sciences, 1105, BA, Amsterdam, The Netherlands
| | - René S Kahn
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Department of Psychiatry, University Medical Center Utrecht Brain Center, Utrecht University, 3584, CG, Utrecht, The Netherlands.,Mental Illness Research Education Clinical, Centers of Excellence, VA, Mental Health, Veterans, Bronx, NY, USA
| | - Lot D de Witte
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Department of Psychiatry, University Medical Center Utrecht Brain Center, Utrecht University, 3584, CG, Utrecht, The Netherlands.,Mental Illness Research Education Clinical, Centers of Excellence, VA, Mental Health, Veterans, Bronx, NY, USA
| |
Collapse
|
23
|
Schwab N, Ju Y, Hazrati LN. Early onset senescence and cognitive impairment in a murine model of repeated mTBI. Acta Neuropathol Commun 2021; 9:82. [PMID: 33964983 PMCID: PMC8106230 DOI: 10.1186/s40478-021-01190-x] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Accepted: 05/03/2021] [Indexed: 12/19/2022] Open
Abstract
Mild traumatic brain injury (mTBI) results in broad neurological symptoms and an increased risk of being diagnosed with a neurodegenerative disease later in life. While the immediate oxidative stress response and post-mortem pathology of the injured brain has been well studied, it remains unclear how early pathogenic changes may drive persistent symptoms and confer susceptibility to neurodegeneration. In this study we have used a mouse model of repeated mTBI (rmTBI) to identify early gene expression changes at 24 h or 7 days post-injury (7 dpi). At 24 h post-injury, gene expression of rmTBI mice shows activation of the DNA damage response (DDR) towards double strand DNA breaks, altered calcium and cell–cell signalling, and inhibition of cell death pathways. By 7 dpi, rmTBI mice had a gene expression signature consistent with induction of cellular senescence, activation of neurodegenerative processes, and inhibition of the DDR. At both timepoints gliosis, microgliosis, and axonal damage were evident in the absence of any gross lesion, and by 7 dpi rmTBI also mice had elevated levels of IL1β, p21, 53BP1, DNA2, and p53, supportive of DNA damage-induced cellular senescence. These gene expression changes reflect establishment of processes usually linked to brain aging and suggests that cellular senescence occurs early and most likely prior to the accumulation of toxic proteins. These molecular changes were accompanied by spatial learning and memory deficits in the Morris water maze. To conclude, we have identified DNA damage-induced cellular senescence as a repercussion of repeated mild traumatic brain injury which correlates with cognitive impairment. Pathways involved in senescence may represent viable treatment targets of post-concussive syndrome. Senescence has been proposed to promote neurodegeneration and appears as an effective target to prevent long-term complications of mTBI, such as chronic traumatic encephalopathy and other related neurodegenerative pathologies.
Collapse
|
24
|
Dang MT, Gonzalez MV, Gaonkar KS, Rathi KS, Young P, Arif S, Zhai L, Alam Z, Devalaraja S, To TKJ, Folkert IW, Raman P, Rokita JL, Martinez D, Taroni JN, Shapiro JA, Greene CS, Savonen C, Mafra F, Hakonarson H, Curran T, Haldar M. Macrophages in SHH subgroup medulloblastoma display dynamic heterogeneity that varies with treatment modality. Cell Rep 2021; 34:108917. [PMID: 33789113 PMCID: PMC10450591 DOI: 10.1016/j.celrep.2021.108917] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 01/13/2021] [Accepted: 03/09/2021] [Indexed: 12/21/2022] Open
Abstract
Tumor-associated macrophages (TAMs) play an important role in tumor immunity and comprise of subsets that have distinct phenotype, function, and ontology. Transcriptomic analyses of human medulloblastoma, the most common malignant pediatric brain cancer, showed that medulloblastomas (MBs) with activated sonic hedgehog signaling (SHH-MB) have significantly more TAMs than other MB subtypes. Therefore, we examined MB-associated TAMs by single-cell RNA sequencing of autochthonous murine SHH-MB at steady state and under two distinct treatment modalities: molecular-targeted inhibitor and radiation. Our analyses reveal significant TAM heterogeneity, identify markers of ontologically distinct TAM subsets, and show the impact of brain microenvironment on the differentiation of tumor-infiltrating monocytes. TAM composition undergoes dramatic changes with treatment and differs significantly between molecular-targeted and radiation therapy. We identify an immunosuppressive monocyte-derived TAM subset that emerges with radiation therapy and demonstrate its role in regulating T cell and neutrophil infiltration in MB.
Collapse
Affiliation(s)
- Mai T Dang
- Division of Neurology, Children's Hospital of Philadelphia, Philadelphia, PA, USA; Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Michael V Gonzalez
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Krutika S Gaonkar
- Center for Data-Driven Discovery in Biomedicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA; Department of Bioinformatics and Health Informatics, Children's Hospital of Philadelphia, Philadelphia, PA, USA; Division of Neurosurgery, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Komal S Rathi
- Center for Data-Driven Discovery in Biomedicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA; Department of Bioinformatics and Health Informatics, Children's Hospital of Philadelphia, Philadelphia, PA, USA; Division of Neurosurgery, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Patricia Young
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Sherjeel Arif
- Center for Data-Driven Discovery in Biomedicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA; Department of Bioinformatics and Health Informatics, Children's Hospital of Philadelphia, Philadelphia, PA, USA; Division of Neurosurgery, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Li Zhai
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Zahidul Alam
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Samir Devalaraja
- Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Tsun Ki Jerrick To
- Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ian W Folkert
- Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Pichai Raman
- Center for Data-Driven Discovery in Biomedicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA; Department of Bioinformatics and Health Informatics, Children's Hospital of Philadelphia, Philadelphia, PA, USA; Division of Neurosurgery, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Jo Lynne Rokita
- Center for Data-Driven Discovery in Biomedicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA; Department of Bioinformatics and Health Informatics, Children's Hospital of Philadelphia, Philadelphia, PA, USA; Division of Neurosurgery, Children's Hospital of Philadelphia, Philadelphia, PA, USA; Alex's Lemonade Stand Foundation Childhood Cancer Data Lab, Philadelphia, PA, USA
| | - Daniel Martinez
- Pathology Core, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Jaclyn N Taroni
- Alex's Lemonade Stand Foundation Childhood Cancer Data Lab, Philadelphia, PA, USA
| | - Joshua A Shapiro
- Alex's Lemonade Stand Foundation Childhood Cancer Data Lab, Philadelphia, PA, USA
| | - Casey S Greene
- Alex's Lemonade Stand Foundation Childhood Cancer Data Lab, Philadelphia, PA, USA; Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Candace Savonen
- Alex's Lemonade Stand Foundation Childhood Cancer Data Lab, Philadelphia, PA, USA
| | - Fernanda Mafra
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Hakon Hakonarson
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, PA, USA; Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Tom Curran
- Children's Research Institute at Mercy Children's Hospital, Kansas City, KS, USA
| | - Malay Haldar
- Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
25
|
Ramaglia V, Rojas O, Naouar I, Gommerman JL. The Ins and Outs of Central Nervous System Inflammation-Lessons Learned from Multiple Sclerosis. Annu Rev Immunol 2021; 39:199-226. [PMID: 33524273 DOI: 10.1146/annurev-immunol-093019-124155] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Multiple sclerosis (MS) is a chronic disease that is characterized by the inappropriate invasion of lymphocytes and monocytes into the central nervous system (CNS), where they orchestrate the demyelination of axons, leading to physical and cognitive disability. There are many reasons immunologists should be interested in MS. Aside from the fact that there is still significant unmet need for patients living with the progressive form of the disease, MS is a case study for how immune cells cross CNS barriers and subsequently interact with specialized tissue parenchymal cells. In this review, we describe the types of immune cells that infiltrate the CNS and then describe interactions between immune cells and glial cells in different types of lesions. Lastly, we provide evidence for CNS-compartmentalized immune cells and speculate on how this impacts disease progression for MS patients.
Collapse
Affiliation(s)
- Valeria Ramaglia
- Department of Immunology, University of Toronto, Ontario M5S 1A8, Canada;
| | - Olga Rojas
- Department of Immunology, University of Toronto, Ontario M5S 1A8, Canada;
| | - Ikbel Naouar
- Department of Immunology, University of Toronto, Ontario M5S 1A8, Canada;
| | | |
Collapse
|
26
|
Spiteri AG, Wishart CL, King NJC. Immovable Object Meets Unstoppable Force? Dialogue Between Resident and Peripheral Myeloid Cells in the Inflamed Brain. Front Immunol 2020; 11:600822. [PMID: 33363542 PMCID: PMC7752943 DOI: 10.3389/fimmu.2020.600822] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 11/05/2020] [Indexed: 12/13/2022] Open
Abstract
Inflammation of the brain parenchyma is characteristic of neurodegenerative, autoimmune, and neuroinflammatory diseases. During this process, microglia, which populate the embryonic brain and become a permanent sentinel myeloid population, are inexorably joined by peripherally derived monocytes, recruited by the central nervous system. These cells can quickly adopt a morphology and immunophenotype similar to microglia. Both microglia and monocytes have been implicated in inducing, enhancing, and/or maintaining immune-mediated pathology and thus disease progression in a number of neuropathologies. For many years, experimental and analytical systems have failed to differentiate resident microglia from peripherally derived myeloid cells accurately. This has impeded our understanding of their precise functions in, and contributions to, these diseases, and hampered the development of novel treatments that could target specific cell subsets. Over the past decade, microglia have been investigated more intensively in the context of neuroimmunological research, fostering the development of more precise experimental systems. In light of our rapidly growing understanding of these cells, we discuss the differential origins of microglia and peripherally derived myeloid cells in the inflamed brain, with an analysis of the problems resolving these cell types phenotypically and morphologically, and highlight recent developments enabling more precise identification.
Collapse
Affiliation(s)
- Alanna G. Spiteri
- Discipline of Pathology, Faculty of Medicine and Health, School of Medical Sciences, The University of Sydney, Sydney, NSW, Australia
- Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia
| | - Claire L. Wishart
- Discipline of Pathology, Faculty of Medicine and Health, School of Medical Sciences, The University of Sydney, Sydney, NSW, Australia
- Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia
| | - Nicholas J. C. King
- Discipline of Pathology, Faculty of Medicine and Health, School of Medical Sciences, The University of Sydney, Sydney, NSW, Australia
- Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia
- Sydney Cytometry Facility, The University of Sydney and Centenary Institute, Sydney, NSW, Australia
- Ramaciotti Facility for Human Systems Biology, The University of Sydney and Centenary Institute, Sydney, NSW, Australia
- Marie Bashir Institute for Infectious Diseases and Biosecurity (MBI), Faculty of Medicine and Health, Sydney Medical School, The University of Sydney, Sydney, NSW, Australia
- Nano Institute, The University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
27
|
Shi L, Rocha M, Zhang W, Jiang M, Li S, Ye Q, Hassan SH, Liu L, Adair MN, Xu J, Luo J, Hu X, Wechsler LR, Chen J, Shi Y. Genome-wide transcriptomic analysis of microglia reveals impaired responses in aged mice after cerebral ischemia. J Cereb Blood Flow Metab 2020; 40:S49-S66. [PMID: 32438860 PMCID: PMC7687039 DOI: 10.1177/0271678x20925655] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 03/24/2020] [Accepted: 04/13/2020] [Indexed: 12/25/2022]
Abstract
Senescence-associated alterations in microglia may have profound impact on cerebral homeostasis and stroke outcomes. However, the lack of a transcriptome-wide comparison between young and aged microglia in the context of ischemia limits our understanding of aging-related mechanisms. Herein, we performed RNA sequencing analysis of microglia purified from cerebral hemispheres of young adult (10-week-old) and aged (18-month-old) mice five days after distal middle cerebral artery occlusion or after sham operation. Considerable transcriptional differences were observed between young and aged microglia in healthy brains, indicating heightened chronic inflammation in aged microglia. Following stroke, the overall transcriptional activation was more robust (>13-fold in the number of genes upregulated) in young microglia than in aged microglia. Gene clusters with functional implications in immune inflammatory responses, immune cell chemotaxis, tissue remodeling, and cell-cell interactions were markedly activated in microglia of young but not aged stroke mice. Consistent with the genomic profiling predictions, post-stroke cerebral infiltration of peripheral immune cells was markedly decreased in aged mice compared to young mice. Moreover, post-ischemic aged microglia demonstrated reduced interaction with neighboring neurons and diminished polarity toward the infarct lesion. These alterations in microglial gene response and behavior may contribute to aging-driven vulnerability and poorer recovery after ischemic stroke.
Collapse
Affiliation(s)
- Ligen Shi
- Department of Neurology, Pittsburgh Institute of Brain Disorders & Recovery and UPMC Stroke Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Marcelo Rocha
- Department of Neurology, Pittsburgh Institute of Brain Disorders & Recovery and UPMC Stroke Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Wenting Zhang
- Department of Neurology, Pittsburgh Institute of Brain Disorders & Recovery and UPMC Stroke Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Ming Jiang
- Department of Neurology, Pittsburgh Institute of Brain Disorders & Recovery and UPMC Stroke Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Sicheng Li
- Department of Neurology, Pittsburgh Institute of Brain Disorders & Recovery and UPMC Stroke Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Qing Ye
- Department of Neurology, Pittsburgh Institute of Brain Disorders & Recovery and UPMC Stroke Institute, University of Pittsburgh, Pittsburgh, PA, USA
- Geriatric Research, Education and Clinical Center, Veterans Affairs Pittsburgh Health Care System, Pittsburgh, PA, USA
| | - Sulaiman H Hassan
- Department of Neurology, Pittsburgh Institute of Brain Disorders & Recovery and UPMC Stroke Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Liqiang Liu
- Department of Neurology, Pittsburgh Institute of Brain Disorders & Recovery and UPMC Stroke Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Maya N Adair
- Department of Neurology, Pittsburgh Institute of Brain Disorders & Recovery and UPMC Stroke Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jing Xu
- Department of Neurology, Pittsburgh Institute of Brain Disorders & Recovery and UPMC Stroke Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jianhua Luo
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Xiaoming Hu
- Department of Neurology, Pittsburgh Institute of Brain Disorders & Recovery and UPMC Stroke Institute, University of Pittsburgh, Pittsburgh, PA, USA
- Geriatric Research, Education and Clinical Center, Veterans Affairs Pittsburgh Health Care System, Pittsburgh, PA, USA
| | - Lawrence R Wechsler
- Department of Neurology, Pittsburgh Institute of Brain Disorders & Recovery and UPMC Stroke Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jun Chen
- Department of Neurology, Pittsburgh Institute of Brain Disorders & Recovery and UPMC Stroke Institute, University of Pittsburgh, Pittsburgh, PA, USA
- Geriatric Research, Education and Clinical Center, Veterans Affairs Pittsburgh Health Care System, Pittsburgh, PA, USA
| | - Yejie Shi
- Department of Neurology, Pittsburgh Institute of Brain Disorders & Recovery and UPMC Stroke Institute, University of Pittsburgh, Pittsburgh, PA, USA
- Geriatric Research, Education and Clinical Center, Veterans Affairs Pittsburgh Health Care System, Pittsburgh, PA, USA
| |
Collapse
|
28
|
Quarta A, Berneman Z, Ponsaerts P. Functional consequences of a close encounter between microglia and brain-infiltrating monocytes during CNS pathology and repair. J Leukoc Biol 2020; 110:89-106. [PMID: 33155726 DOI: 10.1002/jlb.3ru0820-536r] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 10/22/2020] [Accepted: 10/23/2020] [Indexed: 12/12/2022] Open
Abstract
Neuroinflammation is recognized as an important factor contributing to the development and progression of several central nervous system (CNS) disorders. Upon CNS trauma or disease, parenchymal microglia highly proliferate and accumulate in and around the lesion site. In addition, blood-derived monocytes can infiltrate the inflamed CNS in response to cellular damage and/or a compromised blood-brain barrier. Both microglia and infiltrating monocytes are characterized by multiple functional states and can either display highly proinflammatory properties or promote resolution of inflammation and tissue regeneration. Despite sharing some basic immunologic functions, microglia and monocytes display many distinctive features, which ultimately define their contribution to neuropathology. Understanding how the innate immune system participates to brain disease is imperative to identify novel treatment options for CNS inflammatory disorders. In this context, existing and newly developed in vitro platforms for disease modeling are fundamental tools to investigate and modulate microglia and monocyte immune functions within a specific neuropathologic context. In this review, we first briefly summarize the current knowledge on microglia and monocyte ontogenesis, as well as their complex and interconnected contributions to the development of various CNS pathologies. Following the well-recognized concept that both microglia and monocytes can either exert neuroprotective functions or exacerbate tissue damage, we provide a comprehensive overview of cellular models currently available for in vitro study of neuroinflammatory responses. In this context, we highlight how simplified single-cell models may not always correctly recapitulate in vivo biology, hence future research should move toward novel models with higher and multicellular complexity.
Collapse
Affiliation(s)
- Alessandra Quarta
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, Antwerp, Belgium
| | - Zwi Berneman
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, Antwerp, Belgium
| | - Peter Ponsaerts
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, Antwerp, Belgium
| |
Collapse
|
29
|
Moreno-García Á, Bernal-Chico A, Colomer T, Rodríguez-Antigüedad A, Matute C, Mato S. Gene Expression Analysis of Astrocyte and Microglia Endocannabinoid Signaling during Autoimmune Demyelination. Biomolecules 2020; 10:biom10091228. [PMID: 32846891 PMCID: PMC7563448 DOI: 10.3390/biom10091228] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 08/13/2020] [Accepted: 08/20/2020] [Indexed: 12/16/2022] Open
Abstract
The endocannabinoid system is associated with protective effects in multiple sclerosis (MS) that involve attenuated innate immune cell responses. Astrocytes and microglia are modulated by endocannabinoids and participate in the biosynthesis and metabolism of these compounds. However, the role of neuroglial cells as targets and mediators of endocannabinoid signaling in MS is poorly understood. Here we used a microfluidic RT-qPCR screen to assess changes in the expression of the main endocannabinoid signaling genes in astrocytes and microglia purified from female mice during the time-course of experimental autoimmune encephalomyelitis (EAE). We show that astrocytes and microglia upregulate the expression of genes encoding neurotoxic A1 and pro-inflammatory molecules at the acute disease with many of these transcripts remaining elevated during the recovery phase. Both cell populations exhibited an early onset decrease in the gene expression levels of 2-arachidonoylglycerol (2-AG) hydrolytic enzymes that persisted during EAE progression as well as cell-type-specific changes in the transcript levels for genes encoding cannabinoid receptors and molecules involved in anandamide (AEA) signaling. Our results demonstrate that astrocytes and microglia responses to autoimmune demyelination involve alterations in the expression of multiple endocannabinoid signaling-associated genes and suggest that this system may regulate the induction of neurotoxic and pro-inflammatory transcriptional programs in both cell types during MS.
Collapse
Affiliation(s)
- Álvaro Moreno-García
- Department of Neurosciences, University of the Basque Country UPV/EHU, E-48940 Leioa, Spain; (Á.M.-G.); (A.B.-C.); (A.R.-A.); (C.M.)
- Achucarro Basque Center for Neuroscience, E-48940 Leioa, Spain;
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), E-28031 Madrid, Spain
| | - Ana Bernal-Chico
- Department of Neurosciences, University of the Basque Country UPV/EHU, E-48940 Leioa, Spain; (Á.M.-G.); (A.B.-C.); (A.R.-A.); (C.M.)
- Achucarro Basque Center for Neuroscience, E-48940 Leioa, Spain;
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), E-28031 Madrid, Spain
| | - Teresa Colomer
- Achucarro Basque Center for Neuroscience, E-48940 Leioa, Spain;
| | - Alfredo Rodríguez-Antigüedad
- Department of Neurosciences, University of the Basque Country UPV/EHU, E-48940 Leioa, Spain; (Á.M.-G.); (A.B.-C.); (A.R.-A.); (C.M.)
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), E-28031 Madrid, Spain
- Biocruces, Bizkaia, E-48903 Barakaldo, Spain
| | - Carlos Matute
- Department of Neurosciences, University of the Basque Country UPV/EHU, E-48940 Leioa, Spain; (Á.M.-G.); (A.B.-C.); (A.R.-A.); (C.M.)
- Achucarro Basque Center for Neuroscience, E-48940 Leioa, Spain;
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), E-28031 Madrid, Spain
| | - Susana Mato
- Department of Neurosciences, University of the Basque Country UPV/EHU, E-48940 Leioa, Spain; (Á.M.-G.); (A.B.-C.); (A.R.-A.); (C.M.)
- Achucarro Basque Center for Neuroscience, E-48940 Leioa, Spain;
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), E-28031 Madrid, Spain
- Biocruces, Bizkaia, E-48903 Barakaldo, Spain
- Correspondence:
| |
Collapse
|
30
|
Plastini MJ, Desu HL, Brambilla R. Dynamic Responses of Microglia in Animal Models of Multiple Sclerosis. Front Cell Neurosci 2020; 14:269. [PMID: 32973458 PMCID: PMC7468479 DOI: 10.3389/fncel.2020.00269] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Accepted: 07/31/2020] [Indexed: 12/20/2022] Open
Abstract
Microglia play an essential role in maintaining central nervous system (CNS) homeostasis, as well as responding to injury and disease. Most neurological disorders feature microglial activation, a process whereby microglia undergo profound morphological and transcriptional changes aimed at containing CNS damage and promoting repair, but often resulting in overt inflammation that sustains and propagates the neurodegenerative process. This is especially evident in multiple sclerosis (MS), were microglial activation and microglia-driven neuroinflammation are considered key events in the onset, progression, and resolution of the disease. Our understanding of microglial functions in MS has widened exponentially in the last decade by way of new tools and markers to discriminate microglia from other myeloid populations. Consequently, the complex functional and phenotypical diversity of microglia can now be appreciated. This, in combination with a variety of animal models that mimic specific features and processes of MS, has contributed to filling the gap of knowledge in the cascade of events underlying MS pathophysiology. The purpose of this review is to present the most up to date knowledge of the dynamic responses of microglia in the commonly used animal models of MS, specifically the immune-mediated experimental autoimmune encephalomyelitis (EAE) model, and the chemically-induced cuprizone and lysolecithin models. Elucidating the spectrum of microglial functions in these models, from detrimental to protective, is essential to identify emerging targets for therapy and guide drug discovery efforts.
Collapse
Affiliation(s)
- Melanie J Plastini
- The Miami Project To Cure Paralysis, Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL, United States.,The Neuroscience Program, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Haritha L Desu
- The Miami Project To Cure Paralysis, Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL, United States.,The Neuroscience Program, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Roberta Brambilla
- The Miami Project To Cure Paralysis, Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL, United States.,The Neuroscience Program, University of Miami Miller School of Medicine, Miami, FL, United States.,Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark.,BRIDGE-Brain Research Inter-Disciplinary Guided Excellence, Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
31
|
Fatoba O, Itokazu T, Yamashita T. Microglia as therapeutic target in central nervous system disorders. J Pharmacol Sci 2020; 144:102-118. [PMID: 32921391 DOI: 10.1016/j.jphs.2020.07.004] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 05/19/2020] [Accepted: 07/07/2020] [Indexed: 12/17/2022] Open
Abstract
Chronic microglial activation is associated with the pathogenesis of several CNS disorders. Microglia show phenotypic diversity and functional complexity in diseased CNS. Thus, understanding the pathology-specific heterogeneity of microglial behavior is crucial for the future development of microglia-modulating therapy for variety of CNS disorders. This review summarizes up-to-date knowledge on how microglia contribute to CNS homeostasis during development and throughout adulthood. We discuss the heterogeneity of microglial phenotypes in the context of CNS disorders with an emphasis on neurodegenerative diseases, demyelinating diseases, CNS trauma, and epilepsy. We conclude this review with a discussion about the disease-specific heterogeneity of microglial function and how it could be exploited for therapeutic intervention.
Collapse
Affiliation(s)
- Oluwaseun Fatoba
- Department of Molecular Neuroscience, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan; WPI-Immunology Frontier Research Center, Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan.
| | - Takahide Itokazu
- Department of Molecular Neuroscience, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan; Department of Neuro-Medical Science, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Toshihide Yamashita
- Department of Molecular Neuroscience, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan; WPI-Immunology Frontier Research Center, Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan; Department of Neuro-Medical Science, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan.
| |
Collapse
|
32
|
N-Acylethanolamine Acid Amidase contributes to disease progression in a mouse model of multiple sclerosis. Pharmacol Res 2020; 160:105064. [PMID: 32634582 DOI: 10.1016/j.phrs.2020.105064] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Revised: 07/01/2020] [Accepted: 07/02/2020] [Indexed: 12/22/2022]
Abstract
N-Acylethanolamine acid amidase (NAAA) deactivates the endogenous peroxisome proliferator-activated receptor-α (PPAR-α) agonist palmitoylethanolamide (PEA). NAAA-regulated PEA signaling participates in the control of peripheral inflammation, but evidence suggests also a role in the modulation of neuroinflammatory pathologies such as multiple sclerosis (MS). Here we show that disease progression in the mouse experimental autoimmune encephalomyelitis (EAE) model of MS is accompanied by induction of NAAA expression in spinal cord, which in presymptomatic animals is confined to motor neurons and oligodendrocytes but, as EAE progresses, extends to microglia/macrophages and other cell types. As previously reported for NAAA inhibition, genetic NAAA deletion delayed disease onset and attenuated symptom intensity in female EAE mice, suggesting that accrued NAAA expression may contribute to pathology. To further delineate the role of NAAA in EAE, we generated a mouse line that selectively overexpresses the enzyme in macrophages, microglia and other monocyte-derived cells. Non-stimulated alveolar macrophages from these NaaaCD11b+ mice contain higher-than-normal levels of inducible nitric oxide synthase and display an activated morphology. Furthermore, intranasal lipopolysaccharide injections cause greater alveolar leukocyte accumulation in NaaaCD11b+ than in control mice. NaaaCD11b+ mice also display a more aggressive clinical response to EAE induction, compared to their wild-type littermates. The results identify NAAA as a critical control step in EAE pathogenesis, and point to this enzyme as a possible target for the treatment of MS.
Collapse
|
33
|
Melcher V, Graf M, Interlandi M, Moreno N, de Faria FW, Kim SN, Kastrati D, Korbanka S, Alfert A, Gerß J, Meyer zu Hörste G, Hartmann W, Frühwald MC, Dugas M, Schüller U, Hasselblatt M, Albert TK, Kerl K. Macrophage-tumor cell interaction promotes ATRT progression and chemoresistance. Acta Neuropathol 2020; 139:913-936. [PMID: 31848709 DOI: 10.1007/s00401-019-02116-7] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Revised: 12/12/2019] [Accepted: 12/13/2019] [Indexed: 12/12/2022]
Abstract
Atypical teratoid/rhabdoid tumors (ATRT) are known for their heterogeneity concerning pathophysiology and outcome. However, predictive factors within distinct subgroups still need to be uncovered. Using multiplex immunofluorescent staining and single-cell RNA sequencing we unraveled distinct compositions of the immunological tumor microenvironment (TME) across ATRT subgroups. CD68+ cells predominantly infiltrate ATRT-SHH and ATRT-MYC and are a negative prognostic factor for patients' survival. Within the murine ATRT-MYC and ATRT-SHH TME, Cd68+ macrophages are core to intercellular communication with tumor cells. In ATRT-MYC distinct tumor cell phenotypes express macrophage marker genes. These cells are involved in the acquisition of chemotherapy resistance in our relapse xenograft mouse model. In conclusion, the tumor cell-macrophage interaction contributes to ATRT-MYC heterogeneity and potentially to tumor recurrence.
Collapse
|
34
|
Cxcl10 + monocytes define a pathogenic subset in the central nervous system during autoimmune neuroinflammation. Nat Immunol 2020; 21:525-534. [PMID: 32313246 DOI: 10.1038/s41590-020-0661-1] [Citation(s) in RCA: 83] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Accepted: 03/13/2020] [Indexed: 12/29/2022]
Abstract
Multiple sclerosis (MS) is characterized by pathological inflammation that results from the recruitment of lymphoid and myeloid immune cells from the blood into the brain. Due to subset heterogeneity, defining the functional roles of the various cell subsets in acute and chronic stages of MS has been challenging. Here, we used index and transcriptional single-cell sorting to characterize the mononuclear phagocytes that infiltrate the central nervous system from the periphery in mice with experimentally induced autoimmune encephalomyelitis, a model of MS. We identified eight monocyte and three dendritic cell subsets at acute and chronic disease stages in which the defined transcriptional programs pointed toward distinct functions. Monocyte-specific cell ablation identified Cxcl10+ and Saa3+ monocytic subsets with a pathogenic potential. Transfer experiments with different monocyte and precursor subsets indicated that these Cxcl10+ and Saa3+ pathogenic cells were not derived from Ly6C+ monocytes but from early myeloid cell progenitors. These results suggest that blocking specific pathogenic monocytic subsets, including Cxcl10+ and Saa3+ monocytes, could be used for targeted therapeutic interventions.
Collapse
|
35
|
Benmamar-Badel A, Owens T, Wlodarczyk A. Protective Microglial Subset in Development, Aging, and Disease: Lessons From Transcriptomic Studies. Front Immunol 2020; 11:430. [PMID: 32318054 PMCID: PMC7147523 DOI: 10.3389/fimmu.2020.00430] [Citation(s) in RCA: 77] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Accepted: 02/25/2020] [Indexed: 12/25/2022] Open
Abstract
Microglial heterogeneity has been the topic of much discussion in the scientific community. Elucidation of their plasticity and adaptability to disease states triggered early efforts to characterize microglial subsets. Over time, their phenotypes, and later on their homeostatic signature, were revealed, through the use of increasingly advanced transcriptomic techniques. Recently, an increasing number of these "microglial signatures" have been reported in various homeostatic and disease contexts. Remarkably, many of these states show similar overlapping microglial gene expression patterns, both in homeostasis and in disease or injury. In this review, we integrate information from these studies, and we propose a unique subset, for which we introduce a core signature, based on our own research and reports from the literature. We describe that this subset is found in development and in normal aging as well as in diverse diseases. We discuss the functions of this subset as well as how it is induced.
Collapse
Affiliation(s)
- Anouk Benmamar-Badel
- Department of Neurobiology Research, Institute for Molecular Medicine, University of Southern Denmark, Odense, Denmark
- BRIDGE, Brain Research - Inter-Disciplinary Guided Excellence, Odense, Denmark
- Department of Neurology, Slagelse Hospital, Institute of Regional Health Research, Slagelse, Denmark
| | - Trevor Owens
- Department of Neurobiology Research, Institute for Molecular Medicine, University of Southern Denmark, Odense, Denmark
- BRIDGE, Brain Research - Inter-Disciplinary Guided Excellence, Odense, Denmark
| | - Agnieszka Wlodarczyk
- Department of Neurobiology Research, Institute for Molecular Medicine, University of Southern Denmark, Odense, Denmark
- BRIDGE, Brain Research - Inter-Disciplinary Guided Excellence, Odense, Denmark
| |
Collapse
|
36
|
Jager SE, Pallesen LT, Richner M, Harley P, Hore Z, McMahon S, Denk F, Vaegter CB. Changes in the transcriptional fingerprint of satellite glial cells following peripheral nerve injury. Glia 2020; 68:1375-1395. [PMID: 32045043 DOI: 10.1002/glia.23785] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Revised: 01/09/2020] [Accepted: 01/10/2020] [Indexed: 01/13/2023]
Abstract
Satellite glial cells (SGCs) are homeostatic cells enveloping the somata of peripheral sensory and autonomic neurons. A wide variety of neuronal stressors trigger activation of SGCs, contributing to, for example, neuropathic pain through modulation of neuronal activity. However, compared to neurons and other glial cells of the nervous system, SGCs have received modest scientific attention and very little is known about SGC biology, possibly due to the experimental challenges associated with studying them in vivo and in vitro. Utilizing a recently developed method to obtain SGC RNA from dorsal root ganglia (DRG), we took a systematic approach to characterize the SGC transcriptional fingerprint by using next-generation sequencing and, for the first time, obtain an overview of the SGC injury response. Our RNA sequencing data are easily accessible in supporting information in Excel format. They reveal that SGCs are enriched in genes related to the immune system and cell-to-cell communication. Analysis of SGC transcriptional changes in a nerve injury-paradigm reveal a differential response at 3 days versus 14 days postinjury, suggesting dynamic modulation of SGC function over time. Significant downregulation of several genes linked to cholesterol synthesis was observed at both time points. In contrast, regulation of gene clusters linked to the immune system (MHC protein complex and leukocyte migration) was mainly observed after 14 days. Finally, we demonstrate that, after nerve injury, macrophages are in closer physical proximity to both small and large DRG neurons, and that previously reported injury-induced proliferation of SGCs may, in fact, be proliferating macrophages.
Collapse
Affiliation(s)
- Sara E Jager
- Danish Research Institute of Translational Neuroscience (DANDRITE), Nordic-EMBL Partnership for Molecular Medicine, Department of Biomedicine, Aarhus University, Aarhus C, Denmark.,Wolfson Centre for Age-Related Diseases, King's College London, Guy's Campus, London, UK
| | - Lone T Pallesen
- Danish Research Institute of Translational Neuroscience (DANDRITE), Nordic-EMBL Partnership for Molecular Medicine, Department of Biomedicine, Aarhus University, Aarhus C, Denmark
| | - Mette Richner
- Danish Research Institute of Translational Neuroscience (DANDRITE), Nordic-EMBL Partnership for Molecular Medicine, Department of Biomedicine, Aarhus University, Aarhus C, Denmark
| | - Peter Harley
- Wolfson Centre for Age-Related Diseases, King's College London, Guy's Campus, London, UK
| | - Zoe Hore
- Wolfson Centre for Age-Related Diseases, King's College London, Guy's Campus, London, UK
| | - Stephen McMahon
- Wolfson Centre for Age-Related Diseases, King's College London, Guy's Campus, London, UK
| | - Franziska Denk
- Wolfson Centre for Age-Related Diseases, King's College London, Guy's Campus, London, UK
| | - Christian B Vaegter
- Danish Research Institute of Translational Neuroscience (DANDRITE), Nordic-EMBL Partnership for Molecular Medicine, Department of Biomedicine, Aarhus University, Aarhus C, Denmark
| |
Collapse
|
37
|
Abstract
Microglia are resident macrophages of the CNS that are involved in its development, homeostasis and response to infection and damage. Microglial activation is a common feature of neurological disorders, and although in some instances this activation can be damaging, protective and regenerative functions of microglia have been revealed. The most prominent example of the regenerative functions is a role for microglia in supporting regeneration of myelin after injury, a process that is critical for axonal health and relevant to numerous disorders in which loss of myelin integrity is a prevalent feature, such as multiple sclerosis, Alzheimer disease and motor neuron disease. Although drugs that are intended to promote remyelination are entering clinical trials, the mechanisms by which remyelination is controlled and how microglia are involved are not completely understood. In this Review, we discuss work that has identified novel regulators of microglial activation - including molecular drivers, population heterogeneity and turnover - that might influence their pro-remyelination capacity. We also discuss therapeutic targeting of microglia as a potential approach to promoting remyelination.
Collapse
|
38
|
Bell OH, Copland DA, Ward A, Nicholson LB, Lange CAK, Chu CJ, Dick AD. Single Eye mRNA-Seq Reveals Normalisation of the Retinal Microglial Transcriptome Following Acute Inflammation. Front Immunol 2020; 10:3033. [PMID: 31993055 PMCID: PMC6964706 DOI: 10.3389/fimmu.2019.03033] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Accepted: 12/10/2019] [Indexed: 01/07/2023] Open
Abstract
Background: Whether retinal microglia can maintain or restore immune homeostasis during and after inflammation is unclear. We performed single-eye mRNA-sequencing on microglia at different timepoints following a single inflammatory stimulus to characterise their transcriptome during and after resolution of endotoxin-induced uveitis (EIU). Experimental Approach: Cx3cr1 CreER :R26-tdTomato (C57BL/6) male heterozygotes were administered tamoxifen via different regimes at 4-5 weeks of age. Four weeks post-tamoxifen, mice were injected intravitreally with 10 ng lipopolysaccharide (endotoxin induced uveitis, EIU). Six-hundred retinal microglia were obtained by FACS from individual naïve retinas and at 4 h, 18 h, and 2 weeks following EIU induction. Samples were sequenced to a depth of up to 16.7 million reads using the SMART-Seq v4 Ultra Low Input RNA kit. The data was analysed using Partek software and Ingenuity Pathway Analysis. Genes were considered differentially-expressed (DEG) if the FDR step-up p-value was ≤0.05 and the fold-change was ≥±2. Results: Flow cytometric analysis indicates that the Cx3cr1 CreER :R26-tdTomato strain is both sensitive (>95% tagging) and specific (>95% specificity) for microglia when tamoxifen is administered topically to the eye for 3 days. During "early" activation, 613 DEGs were identified. In contrast, 537 DEGs were observed during peak cellular infiltrate and none at 2 weeks, compared to baseline controls (1,069 total unique DEGs). Key marker changes were validated by qPCR, flow cytometry, and fluorescence microscopy. C5AR1 was identified and validated as a robust marker of differentiating microglial subsets during an LPS response. Conclusion: Using EIU to provide a single defined inflammatory stimulus, mRNA-Seq identified acute transcriptional changes in retinal microglia which returned to their original transcriptome after 2 weeks. Yolk-sac derived microglia are capable of restoring their homeostatic state after acute inflammation.
Collapse
Affiliation(s)
- Oliver H Bell
- Academic Unit of Ophthalmology, Translational Health Sciences, University of Bristol, Bristol, United Kingdom
| | - David A Copland
- Academic Unit of Ophthalmology, Translational Health Sciences, University of Bristol, Bristol, United Kingdom
| | - Amy Ward
- Academic Unit of Ophthalmology, Translational Health Sciences, University of Bristol, Bristol, United Kingdom
| | - Lindsay B Nicholson
- Academic Unit of Ophthalmology, Translational Health Sciences, University of Bristol, Bristol, United Kingdom
| | - Clemens A K Lange
- Eye Clinic, Medical Centre, University of Freiburg, Freiburg, Germany.,Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Colin J Chu
- Academic Unit of Ophthalmology, Translational Health Sciences, University of Bristol, Bristol, United Kingdom
| | - Andrew D Dick
- Academic Unit of Ophthalmology, Translational Health Sciences, University of Bristol, Bristol, United Kingdom.,Institute of Ophthalmology and the National Institute for Health Research Biomedical Research Centre, Moorfields Eye Hospital and University College London, London, United Kingdom
| |
Collapse
|
39
|
Convergence between Microglia and Peripheral Macrophages Phenotype during Development and Neuroinflammation. J Neurosci 2019; 40:784-795. [PMID: 31818979 DOI: 10.1523/jneurosci.1523-19.2019] [Citation(s) in RCA: 82] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Revised: 10/18/2019] [Accepted: 10/23/2019] [Indexed: 12/30/2022] Open
Abstract
Differently from other myeloid cells, microglia derive exclusively from precursors originating within the yolk sac and migrate to the CNS under development, without any contribution from fetal liver or postnatal hematopoiesis. Consistent with their unique ontology, microglia may express specific physiological markers, which have been partly described in recent years. Here we wondered whether profiles distinguishing microglia from peripheral macrophages vary with age and under pathology. To this goal, we profiled transcriptomes of microglia throughout the lifespan and included a parallel comparison with peripheral macrophages under physiological and neuroinflammatory settings using age- and sex-matched wild-type and bone marrow chimera mouse models. This comprehensive approach demonstrated that the phenotypic differentiation between microglia and peripheral macrophages is age-dependent and that peripheral macrophages do express some of the most commonly described microglia-specific markers early during development, such as Fcrls, P2ry12, Tmem119, and Trem2. Further, during chronic neuroinflammation CNS-infiltrating macrophages and not peripheral myeloid cells acquire microglial markers, indicating that the CNS niche may instruct peripheral myeloid cells to gain the phenotype and, presumably, the function of the microglia cell. In conclusion, our data provide further evidence about the plasticity of the myeloid cell and suggest caution in the strict definition and application of microglia-specific markers.SIGNIFICANCE STATEMENT Understanding the respective role of microglia and infiltrating monocytes in neuroinflammatory conditions has recently seemed possible by the identification of a specific microglia signature. Here instead we provide evidence that peripheral macrophages may express some of the most commonly described microglia markers at some developmental stages or pathological conditions, in particular during chronic neuroinflammation. Further, our data support the hypothesis about phenotypic plasticity and convergence among distinct myeloid cells so that they may act as a functional unit rather than as different entities, boosting their mutual functions in different phases of disease. This holds relevant implications in the view of the growing use of myeloid cell therapies to treat brain disease in humans.
Collapse
|
40
|
Monaghan KL, Zheng W, Hu G, Wan ECK. Monocytes and Monocyte-Derived Antigen-Presenting Cells Have Distinct Gene Signatures in Experimental Model of Multiple Sclerosis. Front Immunol 2019; 10:2779. [PMID: 31849962 PMCID: PMC6889845 DOI: 10.3389/fimmu.2019.02779] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Accepted: 11/13/2019] [Indexed: 12/11/2022] Open
Abstract
Multiple sclerosis (MS) is a chronic inflammatory disease mediated by a complex interaction between the autoreactive lymphocytes and the effector myeloid cells within the central nervous system (CNS). In a murine model of MS, experimental autoimmune encephalomyelitis (EAE), Ly6Chi monocytes migrate into the CNS and further differentiate into antigen-presenting cells (APCs) during disease progression. Currently, there is no information about gene signatures that can distinguish between monocytes and the monocyte-derived APCs. We developed a surface marker-based strategy to distinguish between these two cell types during the stage of EAE when the clinical symptoms were most severe, and performed transcriptome analysis to compare their gene expression. We report here that the inflammatory CNS environment substantially alters gene expression of monocytes, compared to the monocyte differentiation process within CNS. Monocytes in the CNS express genes that encode proinflammatory cytokines and chemokines, and their expression is mostly maintained when the cells differentiate. Moreover, monocyte-derived APCs express surface markers associated with both dendritic cells and macrophages, and have a significant up-regulation of genes that are critical for antigen presentation. Furthermore, we found that Ccl17, Ccl22, and Ccr7 are expressed in monocyte-derived APCs but not the Ly6Chi monocytes. These findings may shed light on identifying molecular signals that control monocyte differentiation and functions during EAE.
Collapse
Affiliation(s)
- Kelly L. Monaghan
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, WV, United States
| | - Wen Zheng
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, WV, United States
| | - Gangqing Hu
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, WV, United States
- Bioinformatics Core, West Virginia University, Morgantown, WV, United States
| | - Edwin C. K. Wan
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, WV, United States
- Department of Neuroscience, West Virginia University, Morgantown, WV, United States
- Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV, United States
| |
Collapse
|
41
|
DePaula-Silva AB, Gorbea C, Doty DJ, Libbey JE, Sanchez JMS, Hanak TJ, Cazalla D, Fujinami RS. Differential transcriptional profiles identify microglial- and macrophage-specific gene markers expressed during virus-induced neuroinflammation. J Neuroinflammation 2019; 16:152. [PMID: 31325960 PMCID: PMC6642742 DOI: 10.1186/s12974-019-1545-x] [Citation(s) in RCA: 84] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Accepted: 07/10/2019] [Indexed: 01/09/2023] Open
Abstract
Background In the healthy central nervous system (CNS), microglia are found in a homeostatic state and peripheral macrophages are absent from the brain. Microglia play key roles in maintaining CNS homeostasis and acting as first responders to infection and inflammation, and peripheral macrophages infiltrate the CNS during neuroinflammation. Due to their distinct origins and functions, discrimination between these cell populations is essential to the comprehension of neuroinflammatory disorders. Studies comparing the gene profiles of microglia and peripheral macrophages, or macrophages in vitro-derived from bone marrow, under non-infectious conditions of the CNS, have revealed valuable microglial-specific genes. However, studies comparing gene profiles between CNS-infiltrating macrophages and microglia, when both are isolated from the CNS during viral-induced neuroinflammation, are lacking. Methods We isolated, via flow cytometry, microglia and infiltrating macrophages from the brains of Theiler’s murine encephalomyelitis virus-infected C57BL/6 J mice and used RNA-Seq, followed by validation with qPCR, to examine the differential transcriptional profiles of these cells. We utilized primary literature defining subcellular localization to determine whether or not particular proteins extracted from the transcriptional profiles were expressed at the cell surface. The surface expression and cellular specificity of triggering receptor expressed on myeloid cells 1 (TREM-1) protein were examined via flow cytometry. We also examined the immune response gene profile within the transcriptional profiles of these isolated microglia and infiltrating macrophages. Results We have identified and validated new microglial- and macrophage-specific genes, encoding cell surface proteins, expressed at the peak of neuroinflammation. TREM-1 protein was confirmed to be expressed by infiltrating macrophages, not microglia, at the peak of neuroinflammation. We also identified both unique and redundant immune functions, through examination of the immune response gene profiles, of microglia and infiltrating macrophages during neurotropic viral infection. Conclusions The differential expression of cell surface-specific genes during neuroinflammation can potentially be used to discriminate between microglia and macrophages as well as provide a resource that can be further utilized to target and manipulate specific cell responses during neuroinflammation. Electronic supplementary material The online version of this article (10.1186/s12974-019-1545-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Ana Beatriz DePaula-Silva
- Department of Pathology, University of Utah School of Medicine, 15 North Medical Drive East, 2600 EEJMRB, Salt Lake City, UT, 84112, USA
| | - Carlos Gorbea
- Department of Biochemistry, University of Utah, 15 North Medical Drive East, 4100 EEJMRB, Salt Lake City, UT, 84112, USA
| | - Daniel J Doty
- Department of Pathology, University of Utah School of Medicine, 15 North Medical Drive East, 2600 EEJMRB, Salt Lake City, UT, 84112, USA
| | - Jane E Libbey
- Department of Pathology, University of Utah School of Medicine, 15 North Medical Drive East, 2600 EEJMRB, Salt Lake City, UT, 84112, USA
| | - John Michael S Sanchez
- Department of Pathology, University of Utah School of Medicine, 15 North Medical Drive East, 2600 EEJMRB, Salt Lake City, UT, 84112, USA
| | - Tyler J Hanak
- Department of Pathology, University of Utah School of Medicine, 15 North Medical Drive East, 2600 EEJMRB, Salt Lake City, UT, 84112, USA
| | - Demián Cazalla
- Department of Biochemistry, University of Utah, 15 North Medical Drive East, 4100 EEJMRB, Salt Lake City, UT, 84112, USA
| | - Robert S Fujinami
- Department of Pathology, University of Utah School of Medicine, 15 North Medical Drive East, 2600 EEJMRB, Salt Lake City, UT, 84112, USA.
| |
Collapse
|
42
|
Activation of the STING-Dependent Type I Interferon Response Reduces Microglial Reactivity and Neuroinflammation. Neuron 2019; 96:1290-1302.e6. [PMID: 29268096 DOI: 10.1016/j.neuron.2017.11.032] [Citation(s) in RCA: 122] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Revised: 09/29/2017] [Accepted: 11/17/2017] [Indexed: 01/07/2023]
Abstract
Brain aging and neurodegeneration are associated with prominent microglial reactivity and activation of innate immune response pathways, commonly referred to as neuroinflammation. One such pathway, the type I interferon response, recognizes viral or mitochondrial DNA in the cytoplasm via activation of the recently discovered cyclic dinucleotide synthetase cGAS and the cyclic dinucleotide receptor STING. Here we show that the FDA-approved antiviral drug ganciclovir (GCV) induces a type I interferon response independent of its canonical thymidine kinase target. Inhibition of components of the STING pathway, including STING, IRF3, Tbk1, extracellular IFNβ, and the Jak-Stat pathway resulted in reduced activity of GCV and its derivatives. Importantly, functional STING was necessary for GCV to inhibit inflammation in cultured myeloid cells and in a mouse model of multiple sclerosis. Collectively, our findings uncover an unexpected new activity of GCV and identify the STING pathway as a regulator of microglial reactivity and neuroinflammation.
Collapse
|
43
|
Lloyd AF, Davies CL, Holloway RK, Labrak Y, Ireland G, Carradori D, Dillenburg A, Borger E, Soong D, Richardson JC, Kuhlmann T, Williams A, Pollard JW, des Rieux A, Priller J, Miron VE. Central nervous system regeneration is driven by microglia necroptosis and repopulation. Nat Neurosci 2019; 22:1046-1052. [PMID: 31182869 PMCID: PMC6597360 DOI: 10.1038/s41593-019-0418-z] [Citation(s) in RCA: 227] [Impact Index Per Article: 37.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Accepted: 04/29/2019] [Indexed: 12/13/2022]
Abstract
Failed regeneration of CNS myelin contributes to clinical decline in neuroinflammatory and neurodegenerative diseases, for which there is an unmet therapeutic need. Here we reveal that efficient remyelination requires death of proinflammatory microglia followed by repopulation to a pro-regenerative state. We propose that impaired microglia death and/or repopulation may underpin dysregulated microglia activation in neurological diseases, and we reveal therapeutic targets to promote white matter regeneration.
Collapse
Affiliation(s)
- Amy F Lloyd
- Medical Research Council Centre for Reproductive Health, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, UK
| | - Claire L Davies
- Medical Research Council Centre for Reproductive Health, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, UK
| | - Rebecca K Holloway
- Medical Research Council Centre for Reproductive Health, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, UK
| | - Yasmine Labrak
- Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Université Catholique de Louvain, Brussels, Belgium
| | - Graeme Ireland
- Medical Research Council Centre for Reproductive Health, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, UK
| | - Dario Carradori
- Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Université Catholique de Louvain, Brussels, Belgium
| | - Alessandra Dillenburg
- Medical Research Council Centre for Reproductive Health, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, UK
| | - Eva Borger
- Medical Research Council Centre for Regenerative Medicine, The University of Edinburgh, Edinburgh, UK
| | - Daniel Soong
- Medical Research Council Centre for Reproductive Health, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, UK
| | - Jill C Richardson
- Neurosciences Therapeutic Area Unit, GlaxoSmithKline R&D Ltd, Stevenage, UK
- Discovery Research MRL UK, Merck Sharp & Dohme, The London Bioscience Innovation Centre, London, UK
| | - Tanja Kuhlmann
- Institute of Neuropathology, University Hospital Muenster, Muenster, Germany
| | - Anna Williams
- Medical Research Council Centre for Regenerative Medicine, The University of Edinburgh, Edinburgh, UK
| | - Jeffrey W Pollard
- Medical Research Council Centre for Reproductive Health, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, UK
| | - Anne des Rieux
- Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Université Catholique de Louvain, Brussels, Belgium
| | - Josef Priller
- Department of Neuropsychiatry and Laboratory of Molecular Psychiatry, Charité Universitätsmedizin Berlin, Berlin, Germany
- United Kingdom Dementia Research Institute, The University of Edinburgh, Edinburgh, UK
| | - Veronique E Miron
- Medical Research Council Centre for Reproductive Health, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, UK.
| |
Collapse
|
44
|
Nally FK, De Santi C, McCoy CE. Nanomodulation of Macrophages in Multiple Sclerosis. Cells 2019; 8:cells8060543. [PMID: 31195710 PMCID: PMC6628349 DOI: 10.3390/cells8060543] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Revised: 05/31/2019] [Accepted: 06/03/2019] [Indexed: 12/16/2022] Open
Abstract
Multiple Sclerosis (MS) is a chronic demyelinating autoimmune disease primarily affecting young adults. Despite an unclear causal factor, symptoms and pathology arise from the infiltration of peripheral immune cells across the blood brain barrier. Accounting for the largest fraction of this infiltrate, macrophages are functionally heterogeneous innate immune cells capable of adopting either a pro or an anti-inflammatory phenotype, a phenomenon dependent upon cytokine milieu in the CNS. This functional plasticity is of key relevance in MS, where the pro-inflammatory state dominates the early stage, instructing demyelination and axonal loss while the later anti-inflammatory state holds a key role in promoting tissue repair and regeneration in later remission. This review highlights a potential therapeutic benefit of modulating macrophage polarisation to harness the anti-inflammatory and reparative state in MS. Here, we outline the role of macrophages in MS and look at the role of current FDA approved therapeutics in macrophage polarisation. Moreover, we explore the potential of particulate carriers as a novel strategy to manipulate polarisation states in macrophages, whilst examining how optimising macrophage uptake via nanoparticle size and functionalisation could offer a novel therapeutic approach for MS.
Collapse
Affiliation(s)
- Frances K Nally
- Molecular and Cellular Therapeutics, Royal College of Surgeons in Ireland, 123 St Stephen's Green, 2 D02 YN77 Dublin, Ireland.
| | - Chiara De Santi
- Molecular and Cellular Therapeutics, Royal College of Surgeons in Ireland, 123 St Stephen's Green, 2 D02 YN77 Dublin, Ireland.
| | - Claire E McCoy
- Molecular and Cellular Therapeutics, Royal College of Surgeons in Ireland, 123 St Stephen's Green, 2 D02 YN77 Dublin, Ireland.
| |
Collapse
|
45
|
Li Q, Lan X, Han X, Wang J. Expression of Tmem119/ Sall1 and Ccr2/ CD69 in FACS-Sorted Microglia- and Monocyte/Macrophage-Enriched Cell Populations After Intracerebral Hemorrhage. Front Cell Neurosci 2019; 12:520. [PMID: 30687011 PMCID: PMC6333739 DOI: 10.3389/fncel.2018.00520] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Accepted: 12/12/2018] [Indexed: 11/30/2022] Open
Abstract
Activation and polarization of microglia and macrophages are critical events in neuroinflammation and hematoma resolution after intracerebral hemorrhage (ICH). However, distinguishing microglia and monocyte-derived macrophages histologically can be difficult. Although they share most cell surface markers, evidence indicates that the gene regulation and function of these two cell types might be different. Flow cytometry is the gold standard for discriminating between the two cell populations, but it is rarely used in the ICH research field. We developed a flow cytometry protocol to identify and sort microglia and monocyte-derived macrophages from mice that have undergone well-established ICH models induced by collagenase or blood injection. In addition, we combined a recently established magnetic-activated cell separation system that allows eight tissue samples to be assessed together. This protocol can be completed within 5–8 h. Sorted cells are fully preserved and maintain expression of microglia-specific (Tmem119/Sall1) and macrophage-specific (Ccr2/CD69) markers. They retain phagocytic ability, respond to lipopolysaccharide stimulation, and engulf fluorescent latex beads. Thus, this protocol represents a very important tool for researching microglial and monocyte-derived macrophage biologic function after ICH and other brain diseases.
Collapse
Affiliation(s)
- Qian Li
- Department of Anesthesiology and Critical Care Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Xi Lan
- Department of Anesthesiology and Critical Care Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Xiaoning Han
- Department of Anesthesiology and Critical Care Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Jian Wang
- Department of Anesthesiology and Critical Care Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
| |
Collapse
|
46
|
Calovi S, Mut-Arbona P, Sperlágh B. Microglia and the Purinergic Signaling System. Neuroscience 2018; 405:137-147. [PMID: 30582977 DOI: 10.1016/j.neuroscience.2018.12.021] [Citation(s) in RCA: 83] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Revised: 12/13/2018] [Accepted: 12/14/2018] [Indexed: 02/07/2023]
Abstract
Microglia are the main resident immune-competent cell type of the central nervous system (CNS); these cells are highly sensitive to subtle changes in the chemical environment of the brain. Microglia are activated during diverse conditions, such as apoptosis, trauma, inflammation, and infection. The specific activities of microglia result from the confluence of environmental stimuli and the cellular state. It is likely that several signaling systems with different biological functions operate in competition and/or synergy, thus regulating similar microglial behaviors. The purinergic system is one of the fundamental signaling systems that establish microglial behavior in a wide spectrum of conditions. Adenosine tri-phosphate (ATP) belongs to the purinergic signaling system, which includes P2X, P2Y, and P1 receptors, as well as other proteins participating in ATP secretion and extracellular ATP degradation, and molecules that recognize purines as a ligand. In this review, we focus on the latest pre-clinical and basic purinergic system and microglial research, with particular attention to data collected in vivo and ex vivo. This chapter is divided into sections related to microglial ATP release, ATP degradation, and ATP-related actions mediated by P2X and P2Y receptor activation.
Collapse
Affiliation(s)
- Stefano Calovi
- Laboratory of Molecular Pharmacology, Institute of Experimental Medicine, Hungarian Academy of Sciences, Budapest, Hungary; János Szentágothai School of Neurosciences, Semmelweis University School of PhD Studies, Budapest, Hungary
| | - Paula Mut-Arbona
- Laboratory of Molecular Pharmacology, Institute of Experimental Medicine, Hungarian Academy of Sciences, Budapest, Hungary; János Szentágothai School of Neurosciences, Semmelweis University School of PhD Studies, Budapest, Hungary
| | - Beáta Sperlágh
- Laboratory of Molecular Pharmacology, Institute of Experimental Medicine, Hungarian Academy of Sciences, Budapest, Hungary.
| |
Collapse
|
47
|
Microglia in Central Nervous System Inflammation and Multiple Sclerosis Pathology. Trends Mol Med 2018; 25:112-123. [PMID: 30578090 DOI: 10.1016/j.molmed.2018.11.005] [Citation(s) in RCA: 340] [Impact Index Per Article: 48.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Revised: 11/22/2018] [Accepted: 11/26/2018] [Indexed: 02/06/2023]
Abstract
Microglia are the resident macrophages of the central nervous system (CNS). They have important physiological functions in maintaining tissue homeostasis but also contribute to CNS pathology. Microglia respond to changes in the microenvironment, and the resulting reactive phenotype can be very diverse, with both neuroinflammatory and neuroprotective properties, illustrating the plasticity of these cells. Recent progress in understanding the autoimmune neuroinflammatory disease multiple sclerosis (MS) and its animal model experimental autoimmune encephalomyelitis suggests major roles for microglia in the disease, which have drastically changed our view on the function of microglia in MS.
Collapse
|
48
|
Bellizzi MJ, Hammond JW, Li H, Gantz Marker MA, Marker DF, Freeman RS, Gelbard HA. The Mixed-Lineage Kinase Inhibitor URMC-099 Protects Hippocampal Synapses in Experimental Autoimmune Encephalomyelitis. eNeuro 2018; 5:ENEURO.0245-18.2018. [PMID: 30627663 PMCID: PMC6325567 DOI: 10.1523/eneuro.0245-18.2018] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Revised: 10/31/2018] [Accepted: 11/07/2018] [Indexed: 12/04/2022] Open
Abstract
Treatments to stop gray matter degeneration are needed to prevent progressive disability in multiple sclerosis (MS). We tested whether inhibiting mixed-lineage kinases (MLKs), which can drive inflammatory microglial activation and neuronal degeneration, could protect hippocampal synapses in C57BL/6 mice with experimental autoimmune encephalomyelitis (EAE), a disease model that recapitulates the excitatory synaptic injury that occurs widely within the gray matter in MS. URMC-099, a broad spectrum MLK inhibitor with additional activity against leucine-rich repeat kinase 2 (LRRK2) and other kinases, prevented loss of PSD95-positive postsynaptic structures, shifted activated microglia toward a less inflammatory phenotype, and reversed deficits in hippocampal-dependent contextual fear conditioning in EAE mice when administered after the onset of motor symptoms. A narrow spectrum inhibitor designed to be highly selective for MLK3 failed to protect synapses in EAE hippocampi, and could not rescue cultured neurons from trophic deprivation in an in vitro model of MLK-driven neuronal degeneration. These results suggest that URMC-099 may have potential as a neuroprotective treatment in MS and demonstrate that a broad spectrum of inhibition against a combination of MLK and other kinases is more effective in neuroinflammatory disease than selectively targeting a single kinase.
Collapse
MESH Headings
- Animals
- Apoptosis/drug effects
- Apoptosis/genetics
- Calcium-Binding Proteins/metabolism
- Cells, Cultured
- Conditioning, Psychological/drug effects
- Cytokines/genetics
- Cytokines/metabolism
- Disease Models, Animal
- Encephalomyelitis, Autoimmune, Experimental/chemically induced
- Encephalomyelitis, Autoimmune, Experimental/drug therapy
- Encephalomyelitis, Autoimmune, Experimental/pathology
- Encephalomyelitis, Autoimmune, Experimental/physiopathology
- Enzyme Inhibitors/therapeutic use
- Fear/drug effects
- Fear/psychology
- Female
- Hippocampus/pathology
- Male
- Mice
- Mice, Inbred C57BL
- Microfilament Proteins/metabolism
- Myelin-Oligodendrocyte Glycoprotein/toxicity
- Neurons/drug effects
- Neuroprotective Agents/therapeutic use
- Peptide Fragments/toxicity
- Pyridines/therapeutic use
- Pyrroles/therapeutic use
- Superior Cervical Ganglion/cytology
- Synapses/drug effects
Collapse
Affiliation(s)
- Matthew J. Bellizzi
- Center for Neurotherapeutics Discovery, University of Rochester Medical Center, Rochester, NY 14642
- Department of Neurology, University of Rochester Medical Center, Rochester, NY 14642
- Department of Neuroscience, University of Rochester Medical Center, Rochester, NY 14642
| | - Jennetta W. Hammond
- Center for Neurotherapeutics Discovery, University of Rochester Medical Center, Rochester, NY 14642
| | - Herman Li
- Center for Neurotherapeutics Discovery, University of Rochester Medical Center, Rochester, NY 14642
| | - Mary A. Gantz Marker
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, NY 14642
| | - Daniel F. Marker
- Center for Neurotherapeutics Discovery, University of Rochester Medical Center, Rochester, NY 14642
| | - Robert S. Freeman
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, NY 14642
| | - Harris A. Gelbard
- Center for Neurotherapeutics Discovery, University of Rochester Medical Center, Rochester, NY 14642
- Department of Neurology, University of Rochester Medical Center, Rochester, NY 14642
- Department of Neuroscience, University of Rochester Medical Center, Rochester, NY 14642
- Departments of Pediatrics and Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY 14642
| |
Collapse
|
49
|
Hu S, Liu Q, Zang S, Zhang Z, Wang J, Cai X, He X. Microglia Are Derived from Peripheral Blood Mononuclear Cells After Pseudorabies Infection in Mice. Viral Immunol 2018; 31:596-604. [PMID: 30339053 DOI: 10.1089/vim.2018.0064] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Pseudorabies virus (PRV) can spread along the peripheral nerves near the site of infection in the animals, and gradually migrates into the central nervous system, where it leads to the development of brain lesions. The aim of this study was to investigate the dynamics of microglia after PRV inoculation. A mouse model inoculated with PRV was established to study the interactions between PRV and microglia, microglial recruitment, and polarization effects. The mice were subcutaneously inoculated with different doses of PRV-Bartha K61 vaccine strain. The obtained results showed that mouse mortality rates increased with the applied doses of virus, and brain lesions, located in the brain tail and brain stem, were observed in each investigated group. Inflammatory cells were shown to infiltrate through the vasculature into perivascular cuff, and the number of microglia was increased as well. Mouse group treated with a medium infection dose demonstrated a high survival rate while developing serious brain lesions, and therefore, this dose was selected for further experiments. Immunohistochemistry, flow cytometry, and confocal laser scanning microscopy were used to analyze PRV-microglia interactions. After PRV inoculation, proliferating cell nuclear antigen (Pcna) and Iba1 double-positive cells were observed in the brain lesions, together with the activated microglia, suggesting that PRV can induce microglial proliferation and activation. Furthermore, 5-bromo-deoxy-uridine (BrdU) labeling demonstrated that microglial cells did not proliferate in situ and the proliferating cells originated from peripheral blood monocytes, mainly from the inflammatory monocytes (Ly6Chigh). In addition, microglia polarized into both M1 and M2 phenotypes by PRV infection. The results obtained in this study may help understand the development of pseudorabies infection and help improve the treatment, by recruiting and enhancing immune response.
Collapse
Affiliation(s)
- Shouping Hu
- National Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute , Chinese Academy of Agricultural Sciences, Harbin, China
| | - Qiang Liu
- National Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute , Chinese Academy of Agricultural Sciences, Harbin, China
| | - Sufang Zang
- National Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute , Chinese Academy of Agricultural Sciences, Harbin, China
| | - Zhuo Zhang
- National Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute , Chinese Academy of Agricultural Sciences, Harbin, China
| | - Jingfei Wang
- National Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute , Chinese Academy of Agricultural Sciences, Harbin, China
| | - Xuehui Cai
- National Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute , Chinese Academy of Agricultural Sciences, Harbin, China
| | - Xijun He
- National Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute , Chinese Academy of Agricultural Sciences, Harbin, China
| |
Collapse
|
50
|
Peruzzotti-Jametti L, Pluchino S. Targeting Mitochondrial Metabolism in Neuroinflammation: Towards a Therapy for Progressive Multiple Sclerosis. Trends Mol Med 2018; 24:838-855. [DOI: 10.1016/j.molmed.2018.07.007] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Revised: 07/16/2018] [Accepted: 07/17/2018] [Indexed: 02/07/2023]
|